pubmed_id
stringlengths
41
43
abstract
stringlengths
3
18.8k
http://www.ncbi.nlm.nih.gov/pubmed/33436406
1. BMJ Open Respir Res. 2021 Jan;8(1):e000845. doi: 10.1136/bmjresp-2020-000845. Sleep apnoea is a risk factor for severe COVID-19. Strausz S(1)(2)(3), Kiiskinen T(1)(4), Broberg M(1), Ruotsalainen S(1), Koskela J(1)(5), Bachour A(6); FinnGen; Palotie A(1)(5)(7), Palotie T(2)(3), Ripatti S(1)(5)(8), Ollila HM(9)(5)(10). Collaborators: Palotie A, Daly M, Jacob H, Matakidou A, Runz H, John S, Plenge R, McCarthy M, Hunkapiller J, Ehm M, Waterworth D, Fox C, Malarstig A, Klinger K, Call K, Mäkelä T, Kaprio J, Virolainen P, Pulkki K, Kilpi T, Perola M, Partanen J, Pitkäranta A, Kaarteenaho R, Vainio S, Savinainen K, Kosma VM, Kujala U, Tuovila O, Hendolin M, Pakkanen R, Waring J, Riley-Gillis SB, Matakidou A, Runz H, Liu J, Biswas S, Hunkapiller J, Waterworth D, Ehm M, Diogo D, Fox C, Pfizer A, Marshall C, Hu X, Call K, Klinger K, Gossel M, Ripatti S, Schleutker J, Perola M, Arvas M, Carpen O, Hinttala R, Kettunen J, Laaksonen R, Mannermaa A, Paloneva J, Kujala U, Tuovila O, Hendolin M, Pakkanen R, Soininen H, Julkunen V, Remes A, Kälviäinen R, Hiltunen M, Peltola J, Tienari P, Rinne J, Ziemann A, Waring J, Esmaeeli S, Smaoui N, Lehtonen A, Eaton S, Runz H, Lahdenperä S, Biswas S, Michon J, Kerchner G, Hunkapiller J, Bowers N, Teng E, Merck J, Mehta V, Gormley P, Linden K, Whelan C, Xu F, Pulford D, Färkkilä M, Pikkarainen S, Jussila A, Blomster T, Kiviniemi M, Voutilainen M, Georgantas B, Heap G, Waring J, Smaoui N, Rahimov F, Lehtonen A, Usiskin K, Maranville J, Lu T, Bowers N, Oh D, Michon J, Mehta V, Kalpala K, Miller M, Hu X, McCarthy L, Eklund K, Palomäki A, Isomäki P, Pirilä L, Kaipiainen-Seppänen O, Huhtakangas J, Georgantas B, Waring J, Rahimov F, Lertratanakul A, Smaoui N, Close D, Hochfeld M, Bowers N, Michon J, Diogo D, Mehta V, Kalpala K, Bing N, Hu X, Gordillo JE, Mars N, Laitinen T, Pelkonen M, Kauppi P, Kankaanranta H, Harju T, Smaoui N, Close D, GreenbergCelgene S, Chen H, Bowers N, Michon J, Mehta V, Betts J, Ghosh S, Salomaa V, Niiranen T, Juonala M, Metsärinne K, Kähönen M, Junttila J, Laakso M, Pihlajamäki J, Sinisalo J, Taskinen MR, Tuomi T, Laukkanen J, Ben Challis AP, Hunkapiller J, Bowers N, Michon J, Diogo D, Chu A, Mehta V, Parkkinen J, Miller M, Muslin A, Waterworth D, Joensuu H, Meretoja T, Carpen O, Aaltonen L, Auranen A, Karihtala P, Kauppila S, Auvinen P, Elenius K, Popovic R, Waring J, Riley-Gillis B, Lehtonen A, Matakidou A, Schutzman J, Hunkapiller J, Bowers N, Michon J, Mehta V, Loboda A, Chhibber A, Lehtonen H, McDonough S, Crohns M, Kulkarni D, Kaarniranta K, Turunen J, Ollila T, Seitsonen S, Uusitalo H, Aaltonen V, Uusitalo-Järvinen H, Luodonpää M, Hautala N, Runz H, Strauss E, Bowers N, Chen H, Michon J, Podgornaia A, Mehta V, Diogo D, Hoffman J, Tasanen K, Huilaja L, Hannula-Jouppi K, Salmi T, Peltonen S, Koulu L, Harvima I, Kalpala K, Wu Y, Choy D, Michon J, Smaoui N, Rahimov F, Lehtonen A, Waterworth D, Jalanko A, Kajanne R, Lyhs U, Kaunisto M, Davis J, Riley-Gillis B, Quarless D, Petrovski S, Liu J, Chen CY, Bronson P, Yang R, Maranville J, Biswas S, Chang D, Hunkapiller J, Bhangale T, Bowers N, Diogo D, Holzinger E, Gormley P, Wang X, Chen X, Hedman Å, Auro K, Wang C, Xu E, Auge F, Chatelain C, Kurki M, Ripatti S, Daly M, Karjalainen J, Havulinna A, Jalanko A, Palin K, Palta P, Della P, Zhou W, Lemmelä S, Rivas M, Harju J, Palotie A, Lehisto A, Ganna A, Llorens V, Karlsson A, Kristiansson K, Arvas M, Hyvärinen K, Ritari J, Wahlfors T, Koskinen M, Carpen O, Kettunen J, Pylkäs K, Kalaoja M, Karjalainen M, Mantere T, Kangasniemi E, Heikkinen S, Mannermaa A, Laakkonen E, Kononen J, Loukola A, Laiho P, Sistonen T, Kaiharju E, Laukkanen M, Järvensivu E, Lähteenmäki S, Männikkö L, Wong R, Kristiansson K, Mattsson H, Lemmelä S, Hiekkalinna T, Jiménez M, Donner K, Palta P, Pärn K, Nunez-Fontarnau J, Harju J, Kilpeläinen E, Sipilä T, Brein G, Dada A, Awaisa G, Shcherban A, Sipilä T, Laivuori H, Havulinna A, Lemmelä S, Kiiskinen T, Laitinen T, Siirtola H, Tabuenca J, Kallio L, Soini S, Partanen J, Pitkänen K, Vainio S, Savinainen K, Kosma VM, Kuopio T. Author information: (1)Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland. (2)Orthodontics, Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland. (3)Department of Oral and Maxillofacial Diseases, Helsinki University Hospital (HUH), Helsinki, Finland. (4)Finnish Institute for Health and Welfare, Helsinki, Finland. (5)Broad Institute of MIT and Harvard, Cambridge, MA, USA. (6)Sleep Unit, Heart and Lung Center, Helsinki University Hospital (HUH), Helsinki, Finland. (7)Analytic and Translational Genetics Unit (ATGU), Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA. (8)Department of Public Health, University of Helsinki, Helsinki, Finland. (9)Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland [email protected]. (10)Stanford University School of Medicine, Palo Alto, CA, USA. BACKGROUND: Obstructive sleep apnoea (OSA) is associated with higher body mass index (BMI), diabetes, older age and male gender, which are all risk factors for severe COVID-19.We aimed to study if OSA is an independent risk factor for COVID-19 infection or for severe COVID-19. METHODS: OSA diagnosis and COVID-19 infection were extracted from the hospital discharge, causes of death and infectious diseases registries in individuals who participated in the FinnGen study (n=260 405). Severe COVID-19 was defined as COVID-19 requiring hospitalisation. Multivariate logistic regression model was used to examine association. Comorbidities for either COVID-19 or OSA were selected as covariates. We performed a meta-analysis with previous studies. RESULTS: We identified 445 individuals with COVID-19, and 38 (8.5%) of them with OSA of whom 19 out of 91 (20.9%) were hospitalised. OSA associated with COVID-19 hospitalisation independent from age, sex, BMI and comorbidities (p-unadjusted=5.13×10-5, OR-adjusted=2.93 (95% CI 1.02 to 8.39), p-adjusted=0.045). OSA was not associated with the risk of contracting COVID-19 (p=0.25). A meta-analysis of OSA and severe COVID-19 showed association across 15 835 COVID-19 positive controls, and n=1294 patients with OSA with severe COVID-19 (OR=2.37 (95% 1.14 to 4.95), p=0.021). CONCLUSION: Risk for contracting COVID-19 was the same for patients with OSA and those without OSA. In contrast, among COVID-19 positive patients, OSA was associated with higher risk for hospitalisation. Our findings are in line with earlier works and suggest OSA as an independent risk factor for severe COVID-19. © Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. DOI: 10.1136/bmjresp-2020-000845 PMCID: PMC7804843 PMID: 33436406 [Indexed for MEDLINE] Conflict of interest statement: Competing interests: None declared.
http://www.ncbi.nlm.nih.gov/pubmed/33150129
1. Glob Heart. 2020 Sep 22;15(1):64. doi: 10.5334/gh.814. The Relationship of COVID-19 Severity with Cardiovascular Disease and Its Traditional Risk Factors: A Systematic Review and Meta-Analysis. Matsushita K(1)(2), Ding N(1)(2), Kou M(1)(2), Hu X(1)(2), Chen M(1)(2), Gao Y(1)(2), Honda Y(1)(2), Zhao D(1)(2), Dowdy D(1), Mok Y(1)(2), Ishigami J(1)(2), Appel LJ(1)(2). Author information: (1)Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, US. (2)Welch Center for Prevention, Epidemiology, and Clinical Research, US. BACKGROUND: Whether cardiovascular disease (CVD) and its traditional risk factors predict severe coronavirus disease 2019 (COVID-19) is uncertain, in part, because of potential confounding by age and sex. METHODS: We performed a systematic review of studies that explored pre-existing CVD and its traditional risk factors as risk factors of severe COVID-19 (defined as death, acute respiratory distress syndrome, mechanical ventilation, or intensive care unit admission). We searched PubMed and Embase for papers in English with original data (≥10 cases of severe COVID-19). Using random-effects models, we pooled relative risk (RR) estimates and conducted meta-regression analyses. RESULTS: Of the 661 publications identified in our search, 25 papers met our inclusion criteria, with 76,638 COVID-19 patients including 11,766 severe cases. Older age was consistently associated with severe COVID-19 in all eight eligible studies, with RR >~5 in >60-65 versus <50 years. Three studies showed no change in the RR of age after adjusting for covariate(s). In univariate analyses, factors robustly associated with severe COVID-19 were male sex (10 studies; pooled RR = 1.73, [95% CI 1.50-2.01]), hypertension (8 studies; 2.87 [2.09-3.93]), diabetes (9 studies; 3.20 [2.26-4.53]), and CVD (10 studies; 4.97 [3.76-6.58]). RR for male sex was likely to be independent of age. For the other three factors, meta-regression analyses suggested confounding by age. Only four studies reported multivariable analysis, but most of them showed adjusted RR ~2 for hypertension, diabetes, and CVD. No study explored renin-angiotensin system inhibitors as a risk factor for severe COVID-19. CONCLUSIONS: Despite the potential for confounding, these results suggest that hypertension, diabetes, and CVD are independently associated with severe COVID-19 and, together with age and male sex, can be informative for predicting the risk of severe COVID-19. Copyright: © 2020 The Author(s). DOI: 10.5334/gh.814 PMCID: PMC7546112 PMID: 33150129 [Indexed for MEDLINE] Conflict of interest statement: The authors have no competing interests to declare.
http://www.ncbi.nlm.nih.gov/pubmed/33368966
1. HIV Med. 2021 May;22(5):372-378. doi: 10.1111/hiv.13037. Epub 2020 Dec 27. Immune deficiency is a risk factor for severe COVID-19 in people living with HIV. Hoffmann C(1)(2), Casado JL(3), Härter G(4), Vizcarra P(3), Moreno A(3), Cattaneo D(5)(6), Meraviglia P(7), Spinner CD(8), Schabaz F(9), Grunwald S(10), Gervasoni C(5)(7). Author information: (1)ICH Study Center Hamburg, Hamburg, Germany. (2)University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany. (3)Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Madrid, Spain. (4)Medicover Ulm MVZ, Ulm, Germany. (5)Gestione Ambulatoriale Politerapie (GAP) Outpatient Clinic, ASST Fatebenefratelli, Sacco University Hospital, Milan, Italy. (6)Unit of Clinical Pharmacology, ASST Fatebenefratelli, Sacco University Hospital, Milan, Italy. (7)Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Milan, Italy. (8)School of Medicine, Technical University of Munich, University Hospital rechts der Isar, Munich, Germany. (9)MVZ Karlsplatz München, Munich, Germany. (10)Zentrum für Infektiologie Berlin/Prenzlauer Berg, Berlin, Germany. OBJECTIVES: A prior T cell depletion induced by HIV infection may carry deleterious consequences in the current COVID-19 pandemic. Clinical data on patients co-infected with HIV and SARS-CoV-2 are still scarce. METHODS: This multicentre cohort study evaluated risk factors for morbidity and mortality of COVID-19 in people living with HIV (PLWH), infected with SARS-CoV-2 in three countries in different clinical settings. COVID-19 was clinically classified as to be mild-to-moderate or severe. RESULTS: Of 175 patients, 49 (28%) had severe COVID-19 and 7 (4%) patients died. Almost all patients were on antiretroviral therapy (ART) and in 94%, HIV RNA was below 50 copies/mL prior to COVID-19 diagnosis. In the univariate analysis, an age 50 years or older, a CD4+ T cell nadir of < 200/µl, current CD4+ T cells < 350/µl and the presence of at least one comorbidity were significantly associated with severity of COVID-19. No significant association was found for gender, ethnicity, obesity, a detectable HIV RNA, a prior AIDS-defining illness, or tenofovir (which was mainly given as alafenamide) or protease inhibitor use in the current ART. In a multivariate analysis, the only factor associated with risk for severe COVID-19 was a current CD4+ T cell count of < 350/µl (adjusted odds ratio 2.85, 95% confidence interval 1.26-6.44, p=0.01). The only factor associated with mortality was a low CD4 T cell nadir. CONCLUSIONS: In PLWH, immune deficiency is a possible risk factor for severe COVID-19, even in the setting of virological suppression. There is no evidence for a protective effect of PIs or tenofovir alafenamide. © 2020 The Authors. HIV Medicine published by John Wiley & Sons Ltd on behalf of British HIV Association. DOI: 10.1111/hiv.13037 PMID: 33368966 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33163160
1. F1000Res. 2020 Sep 9;9:1107. doi: 10.12688/f1000research.26186.2. eCollection 2020. Predictors of COVID-19 severity: a systematic review and meta-analysis. Mudatsir M(1), Fajar JK(1)(2), Wulandari L(3), Soegiarto G(4), Ilmawan M(5), Purnamasari Y(5), Mahdi BA(4), Jayanto GD(2), Suhendra S(5), Setianingsih YA(6), Hamdani R(7), Suseno DA(8), Agustina K(9), Naim HY(10), Muchlas M(11), Alluza HHD(5), Rosida NA(5), Mayasari M(5), Mustofa M(5), Hartono A(12), Aditya R(5), Prastiwi F(5), Meku FX(5), Sitio M(5), Azmy A(7), Santoso AS(13), Nugroho RA(5), Gersom C(2), Rabaan AA(14), Masyeni S(15), Nainu F(16), Wagner AL(17), Dhama K(18), Harapan H(1)(19). Author information: (1)Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia. (2)Brawijaya Internal Medicine Research Center, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (3)Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia. (4)Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Easy Java, 60286, Indonesia. (5)Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (6)Department of Urology, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60285, Indonesia. (7)Department of Orthopedic Surgery, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (8)Department of Obstetry and Gynecology, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (9)Department of Neurology, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (10)Department of Urology, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (11)Faculty of Animal Science, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (12)Faculty of Medicine, Universitas Negeri Sebelas Maret, Surakarta, Surakarta, 57126, Indonesia. (13)Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia. (14)Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Dhahran, 31311, Saudi Arabia. (15)Department of Internal Medicine, Faculty of Medicine and Health Science, Universitas Warmadewa, Denpasar, Bali, 80235, Indonesia. (16)Faculty of Pharmacy, Hasanuddin University, Makassar, Makassar, 90245, Indonesia. (17)Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA. (18)Division of Pathology, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243 122, India. (19)Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia. Background: The unpredictability of the progression of coronavirus disease 2019 (COVID-19) may be attributed to the low precision of the tools used to predict the prognosis of this disease. Objective: To identify the predictors associated with poor clinical outcomes in patients with COVID-19. Methods: Relevant articles from PubMed, Embase, Cochrane, and Web of Science were searched as of April 5, 2020. The quality of the included papers was appraised using the Newcastle-Ottawa scale (NOS). Data of interest were collected and evaluated for their compatibility for the meta-analysis. Cumulative calculations to determine the correlation and effect estimates were performed using the Z test. Results: In total, 19 papers recording 1,934 mild and 1,644 severe cases of COVID-19 were included. Based on the initial evaluation, 62 potential risk factors were identified for the meta-analysis. Several comorbidities, including chronic respiratory disease, cardiovascular disease, diabetes mellitus, and hypertension were observed more frequent among patients with severe COVID-19 than with the mild ones. Compared to the mild form, severe COVID-19 was associated with symptoms such as dyspnea, anorexia, fatigue, increased respiratory rate, and high systolic blood pressure. Lower levels of lymphocytes and hemoglobin; elevated levels of leukocytes, aspartate aminotransferase, alanine aminotransferase, blood creatinine, blood urea nitrogen, high-sensitivity troponin, creatine kinase, high-sensitivity C-reactive protein, interleukin 6, D-dimer, ferritin, lactate dehydrogenase, and procalcitonin; and a high erythrocyte sedimentation rate were also associated with severe COVID-19. Conclusion: More than 30 risk factors are associated with a higher risk of severe COVID-19. These may serve as useful baseline parameters in the development of prediction tools for COVID-19 prognosis. Copyright: © 2021 Mudatsir M et al. DOI: 10.12688/f1000research.26186.2 PMCID: PMC7607482 PMID: 33163160 [Indexed for MEDLINE] Conflict of interest statement: No competing interests were disclosed.
http://www.ncbi.nlm.nih.gov/pubmed/35137371
1. Adv Exp Med Biol. 2021;1353:115-129. doi: 10.1007/978-3-030-85113-2_7. Risk Factors for COVID-19: Diabetes, Hypertension, and Obesity. Buscemi S(1), Corleo D(2), Randazzo C(2). Author information: (1)Dipartimento di Promozione della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo (Italy), Palermo, Italy. [email protected]. (2)Dipartimento di Promozione della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo (Italy), Palermo, Italy. INTRODUCTION: The recent global pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has involved more than 7 million people worldwide and been associated with more than 400,000 deaths. No clear information is known about all the potential risk factors for COVID-19 or what factors adversely influence its clinical course and mortality. Therefore, we analyzed the role of obesity, type 2 diabetes, and hypertension as risk factors for COVID-19. METHODS: We identified articles for inclusion by searching PubMed and Google Scholar (last accessed 15 June 2020). Retrospective review of literature. Analysis of epidemiological data concerning obesity prevalence and COVID-19 incidence, particularly in Italy and the USA. RESULTS: Data from several retrospective studies of prevalence showed that patients with hypertension, type 2 diabetes, and obesity may have more severe COVID-19, intensive care unit admission, and higher mortality rates, but it is not definitively clear if this is an independent association. In general, the prevalence of obesity in patients with COVID-19 seems to be the same as that of the general population throughout the world; however, obesity seems to be associated with more severe disease and mortality in younger (< 60 years) patients. Similar effects seem to occur in patients with diabetes and/or hypertension but at older ages (> 60 years). In strict connection, it has been proposed that the use of drugs inhibiting angiotensin-converting enzyme 2 (ACE-2) or dipeptidyl dipeptidase 4 (DPP-4) might influence viral activity and disease severity since ACE-2 and DPP-4 receptors mediate SARS-CoV-2 entry into the host cells; however, no evidence exists to date that shows that this may be the case. CONCLUSION: Overall, diabetes, hypertension, and obesity seem to negatively affect the clinical course and disease outcome in patients with COVID-19. However, these data need further confirmation by studies with more accurate data registration. © 2021. The Author(s), under exclusive license to Springer Nature Switzerland AG. DOI: 10.1007/978-3-030-85113-2_7 PMID: 35137371 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32612666
1. Immun Ageing. 2020 Jun 30;17:21. doi: 10.1186/s12979-020-00192-y. eCollection 2020. SARS-CoV-2 disease severity and diabetes: why the connection and what is to be done? Mazucanti CH(1), Egan JM(1). Author information: (1)National Institute on Aging, Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224 USA. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel virus responsible for the current coronavirus disease 2019 (COVID-19) pandemic, has infected over 3.5 million people all over the world since the first case was reported from Wuhan, China 5 months ago. As more epidemiological data regarding COVID-19 patients is acquired, factors that increase the severity of the infection are being identified and reported. One of the most consistent co-morbidities associated with worse outcome in COVID-19 patients is diabetes, along with age and cardiovascular disease. Studies on the association of diabetes with other acute respiratory infections, namely SARS, MERS, and Influenza, outline what seems to be an underlying factor in diabetic patients that makes them more susceptible to complications. In this review we summarize what we think may be the factors driving this pattern between diabetes, aging and poor outcomes in respiratory infections. We also review therapeutic considerations and strategies for treatment of COVID-19 in diabetic patients, and how the additional challenge of this co-morbidity requires attention to glucose homeostasis so as to achieve the best outcomes possible for patients. © The Author(s) 2020. DOI: 10.1186/s12979-020-00192-y PMCID: PMC7325192 PMID: 32612666 Conflict of interest statement: Competing interestsThe authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35124268
1. Clin Gastroenterol Hepatol. 2022 Jul;20(7):1553-1560.e78. doi: 10.1016/j.cgh.2022.01.045. Epub 2022 Feb 3. Mendelian Randomization Analysis Reveals No Causal Relationship Between Nonalcoholic Fatty Liver Disease and Severe COVID-19. Li J(1), Tian A(1), Zhu H(2), Chen L(3), Wen J(4), Liu W(5), Chen P(6). Author information: (1)Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, China. (2)Teaching Department, First Affiliated Hospital of Jilin University, Changchun, China. (3)School of Clinical Medicine, Jilin University, Changchun, China. (4)Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China. (5)Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan. Electronic address: [email protected]. (6)Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, China; Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China. Electronic address: [email protected]. Comment in Clin Gastroenterol Hepatol. 2022 Oct;20(10):2416-2417. doi: 10.1016/j.cgh.2022.03.037. BACKGROUND & AIMS: The coronavirus disease 2019 (COVID-19) pandemic has witnessed more than 4.5 million deaths as of the time of writing. Whether nonalcoholic fatty liver disease (NAFLD) increases the risk for severe COVID-19 remains unclear. We sought to address this question using 2-sample Mendelian randomization (TSMR) analysis approaches in large cohorts. METHODS: We performed large-scale TSMR analyses to examine whether there is a causal relationship between NAFLD, serum alanine aminotransferase, grade of steatosis, NAFLD Activity Score, or fibrosis stage and severe COVID-19. To maximize the power of this analysis, we performed a genome-wide meta-analysis to identify single nucleotide polymorphisms associated with NAFLD. We also examined the impact of 20 major comorbid factors of NAFLD on severe COVID-19. RESULTS: Univariate analysis of the UK Biobank data demonstrated a significant association between NAFLD and severe COVID-19 (odds ratio [OR], 3.06; P = 1.07 × 10-6). However, this association disappeared after demographic and comorbid factors were adjusted (OR, 1.57; P = .09). TSMR study indicated that NAFLD (OR, 0.97; P = .61), alanine aminotransferase level (OR, 1.03; P = .47), grade of steatosis (OR, 1.08; P = .41), NAFLD Activity Score (OR, 1.02; P = .39), and fibrosis stage (OR, 1.01; P = .87) were not associated with severe COVID-19. Among all NAFLD-related comorbid factors, body mass index (OR, 1.73; P = 7.65 × 10-9), waist circumference (OR, 1.76; P = 2.58 × 10-5), and hip circumference (OR, 1.33; P = 7.26 × 10-3) were the only ones demonstrated a causal impact on severe COVID-19. CONCLUSIONS: There is no evidence supporting that NAFLD is a causal risk factor for severe COVID-19. Previous observational associations between NAFLD and COVID-19 are likely attributed to the correlation between NAFLD and obesity. Copyright © 2022 AGA Institute. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.cgh.2022.01.045 PMCID: PMC8812093 PMID: 35124268 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33340043
1. Nephrol Dial Transplant. 2021 Jan 1;36(1):87-94. doi: 10.1093/ndt/gfaa314. Chronic kidney disease is a key risk factor for severe COVID-19: a call to action by the ERA-EDTA. ERA-EDTA Council; ERACODA Working Group. Collaborators: Ortiz A, Cozzolino M, Fliser D, Fouque D, Goumenos D, Massy ZA, Rosenkranz AR, Rychlık I, Soler MJ, Stevens K, Torra R, Tuglular S, Wanner C, Gansevoort RT, Duivenvoorden R, Franssen CFM, Hemmelder MH, Hilbrands LB, Jager KJ, Noordzij M, Vart P, Gansevoort RT. Comment in Nephrol Dial Transplant. 2021 Jan 1;36(1):8-11. doi: 10.1093/ndt/gfaa339. Diabetes, hypertension and cardiovascular disease have been listed as risk factors for severe coronavirus disease 2019 (COVID-19) since the first report of the disease in January 2020. However, this report did not mention chronic kidney disease (CKD) nor did it provide information on the relevance of estimated glomerular filtration rate (eGFR) or albuminuria. As the disease spread across the globe, information on larger populations with greater granularity on risk factors emerged. The recently published OpenSAFELY project analysed factors associated with COVID-19 death in 17 million patients. The picture that arose differs significantly from initial reports. For example, hypertension is not an independent risk factor for COVID-19 death [adjusted hazard ratio (aHR) 0.89], but renal disease very much is. Dialysis (aHR 3.69), organ transplantation (aHR 3.53) and CKD (aHR 2.52 for patients with eGFR <30 mL/min/1.73 m2) represent three of the four comorbidities associated with the highest mortality risk from COVID-19. The risk associated with CKD Stages 4 and 5 is higher than the risk associated with diabetes mellitus (aHR range 1.31-1.95, depending upon glycaemic control) or chronic heart disease (aHR 1.17). In another recent publication, the Global Burden of Disease collaboration identified that worldwide, CKD is the most prevalent risk factor for severe COVID-19. Moreover, the distribution of risk factors for COVID-19 mortality appears to be different in patients with CKD when compared with the general population. The high prevalence of CKD in combination with the elevated risk of mortality from COVID-19 in CKD necessitates urgent action for this group of patients. This article defines essential action points (summarized in Box 1), among which is advocating the inclusion of CKD patients in clinical trials testing the efficacy of drugs and vaccines to prevent severe COVID-19. © The Author(s) 2020. Published by Oxford University Press on behalf of ERA-EDTA. DOI: 10.1093/ndt/gfaa314 PMCID: PMC7771976 PMID: 33340043 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36431276
1. J Clin Med. 2022 Nov 17;11(22):6799. doi: 10.3390/jcm11226799. Concentrations of Soluble Angiotensin Converting Enzyme 2 (sACE2) in Children and Adults with and without COVID-19. Wissing SI(1), Obeid R(2), Rädle-Hurst T(1), Rohrer T(3), Herr C(4), Schöpe J(5), Geisel J(2), Bals R(4)(6), Abdul-Khaliq H(1). Author information: (1)Department of Pediatric Cardiology, Saarland University Hospital, 66421 Homburg, Germany. (2)Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, 66421 Homburg, Germany. (3)Department of Pediatric Endocrinology, Saarland University Hospital, 66421 Homburg, Germany. (4)Department of Internal Medicine V-Pulmonology, Allergology and Critical Care Medicine, Saarland University Hospital, 66421 Homburg, Germany. (5)Institute for Medical Biometry, Epidemiology and Medical Informatics, Saarland University Medical Center, 66421 Homburg, Germany. (6)Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123 Saarbrücken, Germany. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing the coronavirus disease 2019 (COVID-19) pandemic, leads to illness and death. Various risk factors for a severe course, such as higher age, male gender and pre-existing illnesses are known. However, pathophysiological risk factors are largely unclear. Notably, the mild course of disease in children is conspicuous. Angiotensin converting enzyme 2 (ACE2) serves as a receptor for SARS-CoV-2 and is a key enzyme in infection. Differences in the distribution of ACE2 can provide insights into different courses of COVID-19. Our aim was to elucidate the role of ACE2 as a pathophysiological risk factor by measuring soluble ACE2 (sACE2) via ELISA in blood samples (lithium-heparin-plasma or serum) of 367 individuals including children and adults with and without COVID-19. sACE2-levels were compared between the groups according to age and sex. In adults and children with COVID-19, sACE2-concentrations are significantly higher compared to healthy individuals. sACE2-levels increase with age and are lower in children compared to adults with COVID-19. Sex doesn't significantly influence sACE2-concentration. It remains unclear whether sACE2 concentrations increase because of the infection and what factors could influence this response. In conclusion, the increase of sACE2-concentration with age could indicate that ACE2 concentrations mirror increased COVID-19 severity. DOI: 10.3390/jcm11226799 PMCID: PMC9698605 PMID: 36431276 Conflict of interest statement: The authors declare that they have no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/36260598
1. PLoS One. 2022 Oct 19;17(10):e0275101. doi: 10.1371/journal.pone.0275101. eCollection 2022. Observational study of factors associated with morbidity and mortality from COVID-19 in Lebanon, 2020-2021. Nader M(1), Zmerli O(2), Platt DE(3), Hamdan H(1), Hamdash S(4), Tayeh RA(2), Azar J(2), Kadi D(2), Sultan Y(2), Bazarbachi T(4), Karayakoupoglou G(4), Zalloua P(1)(5), Azar E(2). Author information: (1)College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates. (2)Division of Infectious Diseases, Department of Medicine, Saint George Hospital University Medical Center, Beirut, Lebanon. (3)Computational Biology Center, IBM TJ Watson Research Centre, Yorktown Hgts, New York, United States of America. (4)Laboratory Medicine, Haykel Hospital, Tripoli, Lebanon. (5)Harvard T.H. Chan School of Public Health, Boston, Mssachusets, United States of America. BACKGROUND: The COVID-19 pandemic claimed millions of lives worldwide without clear signs of abating despite several mitigation efforts and vaccination campaigns. There have been tremendous interests in understanding the etiology of the disease particularly in what makes it severe and fatal in certain patients. Studies have shown that COVID-19 patients with kidney injury on admission were more likely to develop severe disease, and acute kidney disease was associated with high mortality in COVID-19 hospitalized patients. METHODS: This study investigated 819 COVID-19 patients admitted between January 2020-April 2021 to the COVID-19 ward at a tertiary care center in Lebanon and evaluated their vital signs and biomarkers while probing for two main outcomes: intubation and fatality. Logistic and Cox regressions were performed to investigate the association between clinical and metabolic variables and disease outcomes, mainly intubation and mortality. Times were defined in terms of admission and discharge/fatality for COVID-19, with no other exclusions. RESULTS: Regression analysis revealed that the following are independent risk factors for both intubation and fatality respectively: diabetes (p = 0.021 and p = 0.04), being overweight (p = 0.021 and p = 0.072), chronic kidney disease (p = 0.045 and p = 0.001), and gender (p = 0.016 and p = 0.114). Further, shortness of breath (p<0.001), age (p<0.001) and being overweight (p = 0.014) associated with intubation, while fatality with shortness of breath (p<0.001) in our group of patients. Elevated level of serum creatinine was the highest factor associated with fatality (p = 0.002), while both white blood count (p<0.001) and serum glutamic-oxaloacetic transaminase levels (p<0.001) emerged as independent risk factors for intubation. CONCLUSIONS: Collectively our data show that high creatinine levels were significantly associated with fatality in our COVID-19 study patients, underscoring the importance of kidney function as a main modulator of SARS-CoV-2 morbidity and favor a careful and proactive management of patients with elevated creatinine levels on admission. DOI: 10.1371/journal.pone.0275101 PMCID: PMC9581355 PMID: 36260598 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/33592063
1. PLoS One. 2021 Feb 16;16(2):e0247205. doi: 10.1371/journal.pone.0247205. eCollection 2021. An integrated clinical and genetic model for predicting risk of severe COVID-19: A population-based case-control study. Dite GS(1), Murphy NM(1), Allman R(1). Author information: (1)Genetic Technologies Ltd., Fitzroy, Victoria, Australia. Up to 30% of people who test positive to SARS-CoV-2 will develop severe COVID-19 and require hospitalisation. Age, gender, and comorbidities are known to be risk factors for severe COVID-19 but are generally considered independently without accurate knowledge of the magnitude of their effect on risk, potentially resulting in incorrect risk estimation. There is an urgent need for accurate prediction of the risk of severe COVID-19 for use in workplaces and healthcare settings, and for individual risk management. Clinical risk factors and a panel of 64 single-nucleotide polymorphisms were identified from published data. We used logistic regression to develop a model for severe COVID-19 in 1,582 UK Biobank participants aged 50 years and over who tested positive for the SARS-CoV-2 virus: 1,018 with severe disease and 564 without severe disease. Model discrimination was assessed using the area under the receiver operating characteristic curve (AUC). A model incorporating the SNP score and clinical risk factors (AUC = 0.786; 95% confidence interval = 0.763 to 0.808) had 111% better discrimination of disease severity than a model with just age and gender (AUC = 0.635; 95% confidence interval = 0.607 to 0.662). The effects of age and gender are attenuated by the other risk factors, suggesting that it is those risk factors-not age and gender-that confer risk of severe disease. In the whole UK Biobank, most are at low or only slightly elevated risk, but one-third are at two-fold or more increased risk. We have developed a model that enables accurate prediction of severe COVID-19. Continuing to rely on age and gender alone (or only clinical factors) to determine risk of severe COVID-19 will unnecessarily classify healthy older people as being at high risk and will fail to accurately quantify the increased risk for younger people with comorbidities. DOI: 10.1371/journal.pone.0247205 PMCID: PMC7886160 PMID: 33592063 [Indexed for MEDLINE] Conflict of interest statement: I have read the journal’s policy and the authors of this manuscript have the following competing interests: All authors are employed by Genetic Technologies Limited and have a patent pending (AU_2020901739 – Methods of assessing risk developing a severe response to Coronavirus infection) for the work in this manuscript. A product to predict risk of severe COVID-19 is in development. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
http://www.ncbi.nlm.nih.gov/pubmed/33206661
1. PLoS One. 2020 Nov 18;15(11):e0241541. doi: 10.1371/journal.pone.0241541. eCollection 2020. Factors associated with disease severity and mortality among patients with COVID-19: A systematic review and meta-analysis. Chidambaram V(1), Tun NL(1), Haque WZ(1), Majella MG(2), Sivakumar RK(3), Kumar A(4), Hsu AT(1), Ishak IA(1), Nur AA(1), Ayeh SK(5), Salia EL(6), Zil-E-Ali A(1), Saeed MA(7), Sarena APB(8), Seth B(9), Ahmadzada M(7), Haque EF(10), Neupane P(5), Wang KH(1), Pu TM(1), Ali SMH(11), Arshad MA(12), Wang L(1), Baksh S(1), Karakousis PC(5), Galiatsatos P(9). Author information: (1)Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America. (2)Department of Preventive and Social Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India. (3)Department of Anaesthesia and Intensive Care, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China. (4)Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America. (5)Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America. (6)Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America. (7)Johns Hopkins University, Baltimore, Maryland, United States of America. (8)Bhayangkara Setukpa Hospital, Sukabumi, Indonesia. (9)Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America. (10)Southern Methodist University, Dallas, Texas, United States of America. (11)Fatima Memorial Hospital, Lahore, Pakistan. (12)Nishtar Hospital, Multan, Pakistan. BACKGROUND: Understanding the factors associated with disease severity and mortality in Coronavirus disease (COVID-19) is imperative to effectively triage patients. We performed a systematic review to determine the demographic, clinical, laboratory and radiological factors associated with severity and mortality in COVID-19. METHODS: We searched PubMed, Embase and WHO database for English language articles from inception until May 8, 2020. We included Observational studies with direct comparison of clinical characteristics between a) patients who died and those who survived or b) patients with severe disease and those without severe disease. Data extraction and quality assessment were performed by two authors independently. RESULTS: Among 15680 articles from the literature search, 109 articles were included in the analysis. The risk of mortality was higher in patients with increasing age, male gender (RR 1.45, 95%CI 1.23-1.71), dyspnea (RR 2.55, 95%CI 1.88-2.46), diabetes (RR 1.59, 95%CI 1.41-1.78), hypertension (RR 1.90, 95%CI 1.69-2.15). Congestive heart failure (OR 4.76, 95%CI 1.34-16.97), hilar lymphadenopathy (OR 8.34, 95%CI 2.57-27.08), bilateral lung involvement (OR 4.86, 95%CI 3.19-7.39) and reticular pattern (OR 5.54, 95%CI 1.24-24.67) were associated with severe disease. Clinically relevant cut-offs for leukocytosis(>10.0 x109/L), lymphopenia(< 1.1 x109/L), elevated C-reactive protein(>100mg/L), LDH(>250U/L) and D-dimer(>1mg/L) had higher odds of severe disease and greater risk of mortality. CONCLUSION: Knowledge of the factors associated of disease severity and mortality identified in our study may assist in clinical decision-making and critical-care resource allocation for patients with COVID-19. DOI: 10.1371/journal.pone.0241541 PMCID: PMC7673562 PMID: 33206661 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/34334081
1. Acta Oncol. 2021 Nov;60(11):1459-1465. doi: 10.1080/0284186X.2021.1958005. Epub 2021 Aug 2. Clinical characteristics and factors associated with COVID-19-related death and morbidity among hospitalized patients with cancer: a Swedish cohort study. Ullgren H(1)(2)(3), Camuto A(4), Rosas S(3), Pahnke S(4), Ginman B(4), Enblad G(4), Glimelius I(4), Fransson P(1), Friesland S(3)(5), Liu LL(3)(5). Author information: (1)Department of Nursing, Umeå University, Umeå, Sweden. (2)Regional Cancer Center, Stockholm-Gotland, Sweden. (3)Theme Cancer, Karolinska University Hospital, Stockholm, Sweden. (4)Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden. (5)Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden. INTRODUCTION: Cancer patients are considered to have a higher risk of dying and developing severe Coronavirus Disease 2019 (COVID-19). To date, there are few studies including co-morbidities and sociodemographic factors when investigating the outcome of COVID-19 in a cohort of cancer patients. In this study, we analyzed cancer patients that have been hospitalized due to COVID-19 during the first wave of the pandemic in Sweden to investigate the impact of COVID-19 on mortality and morbidity. PATIENTS AND METHODS: We retrospectively collected data on all patients with cancer that were hospitalized due to COVID-19-related symptoms at Uppsala University Hospital and Karolinska University Hospital between 1 March and 31 August 2020. The primary endpoint was COVID-19-related death and the secondary endpoint was to describe COVID-19 severity, defined as symptom severity (grades 0-4) and length of stay (LOS) at the university hospitals. RESULTS: In total, 193 patients were included among which 31% died due to COVID-19 and 8% died of other causes. In a multivariable analysis, older age >70 (OR 3.6; 95% CI [1.8-7.3], p < 0.001) and male gender (OR 2.8 [1.4-5.8], p = 0.005) were factors associated with higher likelihood of COVID-19-related death. Several comorbidities ≥2 (OR 5.4 [2.0-14.3], p = 0.001) was independently associated with COVID-19 severity. Treatment with chemotherapy within 90 days prior to COVID-19 diagnosis were not associated with COVID-19-related death or severity. CONCLUSION: Factors associated with higher likelihood of COVID-19-related death were older age and male gender. More severe COVID-19 symptoms were seen in patients with multiple comorbidities. We did not see any associations between COVID-19-related death or severity and recent treatment including chemotherapy. In summary, this supports a thorough assessment regarding potential risks with COVID-19 infection in patients with cancer, with a combination of individual risk factors in addition to cancer treatments. DOI: 10.1080/0284186X.2021.1958005 PMID: 34334081 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34935038
1. Pediatrics. 2022 Jan 1;149(1):e2021053418. doi: 10.1542/peds.2021-053418. Epub 2021 Dec 22. Risk Factors for Severe COVID-19 in Children. Woodruff RC(1)(2), Campbell AP(1), Taylor CA(1), Chai SJ(2)(3)(4), Kawasaki B(5), Meek J(6), Anderson EJ(7)(8)(9), Weigel A(10), Monroe ML(11), Reeg L(12), Bye E(13), Sosin DM(14)(15), Muse A(16), Bennett NM(17), Billing LM(18), Sutton M(19), Talbot HK(20), McCaffrey K(21), Pham H(1), Patel K(1)(22), Whitaker M(1), L McMorrow M(1)(2), P Havers F(1)(2). Author information: (1)CDC COVID-19 Response Team. (2)US Public Health Service Commissioned Corps, Rockville, Maryland. (3)Division of State and Local Readiness, Center for Preparedness and Response, Centers for Disease Control and Prevention, Atlanta, Georgia. (4)California Emerging Infections Program, Oakland, California. (5)Colorado Department of Public Health and Environment, Denver, Colorado. (6)Connecticut Emerging Infections Program, Yale School of Public Health, New Haven, Connecticut. (7)Departments of Medicine and Pediatrics, Emory School of Medicine, Atlanta, Georgia. (8)Georgia Emerging Infections Program, Georgia Department of Public Health, Atlanta, Georgia. (9)Atlanta Veterans Affairs Medical Center, Atlanta, Georgia. (10)Iowa Department of Public Health, Des Moines, Iowa. (11)Maryland Department of Health, Baltimore, Maryland. (12)Michigan Department of Health and Human Services, Lansing, Michigan. (13)Minnesota Department of Health, St Paul, Minnesota. (14)New Mexico Emerging Infections Program, Santa Fe, New Mexico. (15)New Mexico Department of Health, Santa Fe, New Mexico. (16)New York State Department of Health, Albany, New York. (17)University of Rochester School of Medicine and Dentistry, Rochester, New York. (18)Ohio Department of Health, Columbus, Ohio. (19)Public Health Division, Oregon Health Authority, Portland, Oregon. (20)Vanderbilt University Medical Center, Nashville, Tennesee. (21)Utah Department of Health, Salt Lake City, Utah. (22)General Dynamics Information Technology, Atlanta, Georgia. OBJECTIVES: Describe population-based rates and risk factors for severe coronavirus disease 2019 (COVID-19) (ie, ICU admission, invasive mechanical ventilation, or death) among hospitalized children. METHODS: During March 2020 to May 2021, the COVID-19-Associated Hospitalization Surveillance Network identified 3106 children hospitalized with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 infection in 14 states. Among 2293 children primarily admitted for COVID-19, multivariable generalized estimating equations generated adjusted risk ratios (aRRs) and 95% confidence intervals (CIs) of the associations between demographic and medical characteristics abstracted from medical records and severe COVID-19. We calculated age-adjusted cumulative population-based rates of severe COVID-19 among all children. RESULTS: Approximately 30% of hospitalized children had severe COVID-19; 0.5% died during hospitalization. Among hospitalized children aged <2 years, chronic lung disease (aRR: 2.2; 95% CI: 1.1-4.3), neurologic disorders (aRR: 2.0; 95% CI: 1.5‒2.6), cardiovascular disease (aRR: 1.7; 95% CI: 1.2‒2.3), prematurity (aRR: 1.6; 95% CI: 1.1‒2.2), and airway abnormality (aRR: 1.6; 95% CI: 1.1‒2.2) were associated with severe COVID-19. Among hospitalized children aged 2 to 17 years, feeding tube dependence (aRR: 2.0; 95% CI: 1.5‒2.5), diabetes mellitus (aRR: 1.9; 95% CI: 1.6‒2.3) and obesity (aRR: 1.2; 95% CI: 1.0‒1.4) were associated with severe COVID-19. Severe COVID-19 occurred among 12.0 per 100 000 children overall and was highest among infants, Hispanic children, and non-Hispanic Black children. CONCLUSIONS: Results identify children at potentially higher risk of severe COVID-19 who may benefit from prevention efforts, including vaccination. Rates establish a baseline for monitoring changes in pediatric illness severity after increased availability of COVID-19 vaccines and the emergence of new variants. Copyright © 2022 by the American Academy of Pediatrics. DOI: 10.1542/peds.2021-053418 PMCID: PMC9213563 PMID: 34935038 [Indexed for MEDLINE] Conflict of interest statement: POTENTIAL CONFLICT OF INTEREST: Mr Meek and Dr Sutton report receiving funding from the Centers for Disease Control and Prevention (CDC) Emerging Infections Program cooperative agreement. Ms Reeg and Ms Billing report receiving a CDC federal grant from the Council of State and Territorial Epidemiologists. Ms Billing reports receiving Epidemiology and Laboratory Capacity grant funding from CDC to support vaccine preventable disease epidemiology staffing and additionally report receiving Immunizations and Vaccines for Children grant funding from CDC. Dr Anderson has consulted for Pfizer, Sanofi Pasteur, Janssen, and Medscape, and his institution receives funds to conduct clinical research unrelated to this article from MedImmune, Regeneron, PaxVax, Pfizer, GSK, Merck, Sanofi Pasteur, Janssen, and Micron. He also serves on a safety monitoring board for Kentucky BioProcessing, Inc. and Sanofi Pasteur. His institution has also received funding from National Institutes of Health to conduct clinical trials of Moderna and Janssen COVID-19 vaccines.
http://www.ncbi.nlm.nih.gov/pubmed/32588943
1. Diabetes Metab Res Rev. 2021 Feb;37(2):e3377. doi: 10.1002/dmrr.3377. Epub 2020 Jul 20. Obesity and diabetes as high-risk factors for severe coronavirus disease 2019 (Covid-19). Zhou Y(1), Chi J(1), Lv W(1), Wang Y(1). Author information: (1)Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China. The outbreak of the coronavirus disease 2019 (Covid-19) has become an evolving worldwide health crisis. With the rising prevalence of obesity and diabetes has come an increasing awareness of their impacts on infectious diseases, including increased risk for various infections, post-infection complications and mortality from critical infections. Although epidemiological and clinical characteristics of Covid-19 have been constantly reported, no article has systematically illustrated the role of obesity and diabetes in Covid-19, or how Covid-19 affects obesity and diabetes, or special treatment in these at-risk populations. Here, we present a synthesis of the recent advances in our understanding of the relationships between obesity, diabetes and Covid-19 along with the underlying mechanisms, and provide special treatment guidance for these at-risk populations. © 2020 John Wiley & Sons Ltd. DOI: 10.1002/dmrr.3377 PMCID: PMC7361201 PMID: 32588943 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no potential conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/34165257
1. HLA. 2021 Aug;98(2):122-131. doi: 10.1111/tan.14349. Epub 2021 Jun 29. HLA-A homozygosis is associated with susceptibility to COVID-19. De Marco R(1), Faria TC(1), Mine KL(1), Cristelli M(2), Medina-Pestana JO(2), Tedesco-Silva H(2), Gerbase-DeLima M(1). Author information: (1)Instituto de Imunogenética, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil. (2)Nephrology Division, Hospital do Rim, Universidade Federal de São Paulo, São Paulo, Brazil. The purpose of this single center retrospective study was to investigate the relationship between HLA and ABO polymorphisms and COVID-19 susceptibility and severity in kidney transplant recipients. It included 720 recipients who had COVID-19 and 1680 controls composed by recipients in follow-up who did not contact the transplantation center for COVID-19 symptoms, up to the moment of their inclusion in the study. HLA-A, -B, and -DRB1 allele groups and ABO frequencies were compared between recipients with COVID-19 (all cases, or separately mild/moderate and severe disease) and controls. The HLA association study was conducted in two case-control series and only associations that showed a p-value <0.05 in both series were considered. No HLA association regarding COVID-19 occurrence or severity met this criterion. Homozygosity at HLA-A locus was associated with COVID-19 susceptibility (odds ratio 1.4) but not severity. Blood groups A and O were associated with susceptibility and resistance to COVID-19, respectively. COVID-19 severity was associated only with older age and cardiac disease, in a multivariate analysis. We conclude that an influence of HLA on COVID-19 susceptibility is supported by the association with homozygosity at HLA-A locus but that there is no evidence for a role of any particular HLA-A, -B, or -DRB1 polymorphism. Thus, we suggest that what matters is the overall capability of an individual's HLA molecules to present SARS-CoV-2 peptides to T cells, a factor that might have a great influence on the breadth of the immune response. © 2021 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd. DOI: 10.1111/tan.14349 PMCID: PMC8446943 PMID: 34165257 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no potential conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32167524
1. JAMA Intern Med. 2020 Jul 1;180(7):934-943. doi: 10.1001/jamainternmed.2020.0994. Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease 2019 Pneumonia in Wuhan, China. Wu C(1)(2)(3), Chen X(3), Cai Y(2), Xia J(4), Zhou X(2), Xu S(2), Huang H(4), Zhang L(4), Zhou X(4), Du C(1), Zhang Y(3), Song J(3), Wang S(3), Chao Y(3), Yang Z(5), Xu J(6), Zhou X(7), Chen D(8), Xiong W(9), Xu L(10), Zhou F(1), Jiang J(3), Bai C(3)(11), Zheng J(12), Song Y(1)(3)(11)(13). Author information: (1)Department of Pulmonary Medicine, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China. (2)Infection Division, Wuhan Jinyintan Hospital, Wuhan, China. (3)Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China. (4)Tuberculosis and Respiratory Department, Wuhan Jinyintan Hospital, Wuhan, China. (5)Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (6)Department of Infectious Diseases, Fengxian Guhua Hospital, Shanghai, China. (7)Department of Pulmonary Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (8)Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (9)Department of Respiratory Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (10)Department of Emergency Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China. (11)Shanghai Respiratory Research Institute, Shanghai, China. (12)Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. (13)National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China. Erratum in JAMA Intern Med. 2020 Jul 1;180(7):1031. doi: 10.1001/jamainternmed.2020.1429. Comment in Signal Transduct Target Ther. 2020 Apr 28;5(1):57. doi: 10.1038/s41392-020-0158-2. JAMA Intern Med. 2020 Jul 1;180(7):1028-1029. doi: 10.1001/jamainternmed.2020.1576. J Intern Med. 2021 Feb;289(2):259-263. doi: 10.1111/joim.13145. JAMA Intern Med. 2021 Jan 1;181(1):140. doi: 10.1001/jamainternmed.2020.2444. JAMA Intern Med. 2021 Jan 1;181(1):139-140. doi: 10.1001/jamainternmed.2020.2438. JAMA Intern Med. 2020 Dec 1;180(12):1715-1716. doi: 10.1001/jamainternmed.2020.3532. IMPORTANCE: Coronavirus disease 2019 (COVID-19) is an emerging infectious disease that was first reported in Wuhan, China, and has subsequently spread worldwide. Risk factors for the clinical outcomes of COVID-19 pneumonia have not yet been well delineated. OBJECTIVE: To describe the clinical characteristics and outcomes in patients with COVID-19 pneumonia who developed acute respiratory distress syndrome (ARDS) or died. DESIGN, SETTING, AND PARTICIPANTS: Retrospective cohort study of 201 patients with confirmed COVID-19 pneumonia admitted to Wuhan Jinyintan Hospital in China between December 25, 2019, and January 26, 2020. The final date of follow-up was February 13, 2020. EXPOSURES: Confirmed COVID-19 pneumonia. MAIN OUTCOMES AND MEASURES: The development of ARDS and death. Epidemiological, demographic, clinical, laboratory, management, treatment, and outcome data were also collected and analyzed. RESULTS: Of 201 patients, the median age was 51 years (interquartile range, 43-60 years), and 128 (63.7%) patients were men. Eighty-four patients (41.8%) developed ARDS, and of those 84 patients, 44 (52.4%) died. In those who developed ARDS, compared with those who did not, more patients presented with dyspnea (50 of 84 [59.5%] patients and 30 of 117 [25.6%] patients, respectively [difference, 33.9%; 95% CI, 19.7%-48.1%]) and had comorbidities such as hypertension (23 of 84 [27.4%] patients and 16 of 117 [13.7%] patients, respectively [difference, 13.7%; 95% CI, 1.3%-26.1%]) and diabetes (16 of 84 [19.0%] patients and 6 of 117 [5.1%] patients, respectively [difference, 13.9%; 95% CI, 3.6%-24.2%]). In bivariate Cox regression analysis, risk factors associated with the development of ARDS and progression from ARDS to death included older age (hazard ratio [HR], 3.26; 95% CI 2.08-5.11; and HR, 6.17; 95% CI, 3.26-11.67, respectively), neutrophilia (HR, 1.14; 95% CI, 1.09-1.19; and HR, 1.08; 95% CI, 1.01-1.17, respectively), and organ and coagulation dysfunction (eg, higher lactate dehydrogenase [HR, 1.61; 95% CI, 1.44-1.79; and HR, 1.30; 95% CI, 1.11-1.52, respectively] and D-dimer [HR, 1.03; 95% CI, 1.01-1.04; and HR, 1.02; 95% CI, 1.01-1.04, respectively]). High fever (≥39 °C) was associated with higher likelihood of ARDS development (HR, 1.77; 95% CI, 1.11-2.84) and lower likelihood of death (HR, 0.41; 95% CI, 0.21-0.82). Among patients with ARDS, treatment with methylprednisolone decreased the risk of death (HR, 0.38; 95% CI, 0.20-0.72). CONCLUSIONS AND RELEVANCE: Older age was associated with greater risk of development of ARDS and death likely owing to less rigorous immune response. Although high fever was associated with the development of ARDS, it was also associated with better outcomes among patients with ARDS. Moreover, treatment with methylprednisolone may be beneficial for patients who develop ARDS. DOI: 10.1001/jamainternmed.2020.0994 PMCID: PMC7070509 PMID: 32167524 [Indexed for MEDLINE] Conflict of interest statement: Conflict of Interest Disclosures: None reported.
http://www.ncbi.nlm.nih.gov/pubmed/35132841
1. J Korean Med Sci. 2022 Feb 7;37(5):e35. doi: 10.3346/jkms.2022.37.e35. Risk Factors for Severe COVID-19 in Children: A Systematic Review and Meta-Analysis. Choi JH(1), Choi SH(2), Yun KW(3). Author information: (1)Department of Pediatrics, Jeju National University School of Medicine, Jeju, Korea. (2)Department of Pediatrics, Pusan National University Hospital, Busan, Korea. (3)Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea. [email protected]. BACKGROUND: Coronavirus disease 2019 (COVID-19) has been the most important global issue since December 2019. Although the clinical course of COVID-19 is known to be milder in children than in adults, associated hospitalizations among children have increased since the emergence of contagious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and the achievement of a high vaccination rate in adults. Considering these global and domestic situations, we believe that risk stratification in children with COVID-19 is urgently needed for decision making regarding hospitalization priority in children infected with SARS-CoV-2 and vaccination priority against COVID-19. METHODS: This systematic review and meta-analysis was performed by comprehensively searching the PubMed, EMBASE, Scopus and KoreaMed databases through August 25, 2021. The criteria for enrollment were "severe COVID-19" as poor outcomes (intensive care unit admission, invasive mechanical ventilation, and/or death) and underlying comorbidities before SARS-CoV-2 infection. RESULTS: Among 872 screened studies, 17 articles were included in the systematic review, and 10 articles were included in the meta-analysis. Neonate (risk ratio [RR], 2.69; 95% confidence interval [CI], 1.83-3.97), prematurity in young infants (RR, 2.00; 95% CI, 1.63-2.46), obesity (RR, 1.43; 95% CI, 1.24-1.64), diabetes (RR, 2.26; 95% CI, 1.95-2.62), chronic lung disease (RR, 2.62; 95% CI, 1.71-4.00), heart disease (RR, 1.82; 95% CI, 1.58-2.09), neurologic disease (RR, 1.18; 95% CI, 1.05-1.33), and immunocompromised status (RR, 1.44; 95% CI, 1.01-2.04) were significant risk factors for severe COVID-19 in children. In the subgroup analysis, age younger than 3 months (RR, 0.26; 95% CI, 0.11-0.66), asthma (RR, 1.08; 95% CI, 0.98-1.20), and neurodevelopmental disorders (RR, 0.88; 95% CI, 0.75-1.04) were not risk factors for severe COVID-19. CONCLUSION: Children with comorbidities such as obesity, diabetes, heart disease, chronic lung diseases other than asthma, seizure disorders, and an immunocompromised status had a high prevalence of severe COVID-19. Neonate and premature infants had a high risk of severe COVID-19. Defining the high-risk group for severe COVID-19 could help to guide hospital admission and priority for vaccination against SARS-CoV-2. © 2022 The Korean Academy of Medical Sciences. DOI: 10.3346/jkms.2022.37.e35 PMCID: PMC8822112 PMID: 35132841 [Indexed for MEDLINE] Conflict of interest statement: The authors have no potential conflicts of interest to disclose.
http://www.ncbi.nlm.nih.gov/pubmed/33063089
1. Cardiovasc Res. 2020 Dec 1;116(14):2197-2206. doi: 10.1093/cvr/cvaa284. Higher mortality of COVID-19 in males: sex differences in immune response and cardiovascular comorbidities. Bienvenu LA(1)(2)(3), Noonan J(1)(3)(4)(5), Wang X(1)(2)(3)(6), Peter K(1)(3)(4)(6). Author information: (1)Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, VIC 3004, Australia. (2)Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. (3)Department of Cardiometabolic Health, University of Melbourne, VIC, Australia. (4)Deparment of Immunology, Monash University, Melbourne, VIC, Australia. (5)Centre for Immunobiology, College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK. (6)Department of Medicine, Monash University, Melbourne, VIC, Australia. The high mortality rate of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection is a critical concern of the coronavirus disease 2019 (COVID-19) pandemic. Strikingly, men account for the majority of COVID-19 deaths, with current figures ranging from 59% to 75% of total mortality. However, despite clear implications in relation to COVID-19 mortality, most research has not considered sex as a critical factor in data analysis. Here, we highlight fundamental biological differences that exist between males and females, and how these may make significant contributions to the male-biased COVID-19 mortality. We present preclinical evidence identifying the influence of biological sex on the expression and regulation of angiotensin-converting enzyme 2 (ACE2), which is the main receptor used by SARS-CoV-2 to enter cells. However, we note that there is a lack of reports showing that sexual dimorphism of ACE2 expression exists and is of functional relevance in humans. In contrast, there is strong evidence, especially in the context of viral infections, that sexual dimorphism plays a central role in the genetic and hormonal regulation of immune responses, both of the innate and the adaptive immune system. We review evidence supporting that ineffective anti-SARS-CoV-2 responses, coupled with a predisposition for inappropriate hyperinflammatory responses, could provide a biological explanation for the male bias in COVID-19 mortality. A prominent finding in COVID-19 is the increased risk of death with pre-existing cardiovascular comorbidities, such as hypertension, obesity, and age. We contextualize how important features of sexual dimorphism and inflammation in COVID-19 may exhibit a reciprocal relationship with comorbidities, and explain their increased mortality risk. Ultimately, we demonstrate that biological sex is a fundamental variable of critical relevance to our mechanistic understanding of SARS-CoV-2 infection and the pursuit of effective COVID-19 preventative and therapeutic strategies. Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2020. For permissions, please email: [email protected]. DOI: 10.1093/cvr/cvaa284 PMCID: PMC7665363 PMID: 33063089 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33661992
1. PLoS One. 2021 Mar 4;16(3):e0247461. doi: 10.1371/journal.pone.0247461. eCollection 2021. Population risk factors for severe disease and mortality in COVID-19: A global systematic review and meta-analysis. Booth A(1), Reed AB(1), Ponzo S(1), Yassaee A(1), Aral M(1), Plans D(1)(2), Labrique A(3), Mohan D(3). Author information: (1)Huma Therapeutics Limited, London, United Kingdom. (2)INDEX Group, Department of Science, Innovation, Technology, and Entrepreneurship, University of Exeter, Exeter, United Kingdom. (3)Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America. AIM: COVID-19 clinical presentation is heterogeneous, ranging from asymptomatic to severe cases. While there are a number of early publications relating to risk factors for COVID-19 infection, low sample size and heterogeneity in study design impacted consolidation of early findings. There is a pressing need to identify the factors which predispose patients to severe cases of COVID-19. For rapid and widespread risk stratification, these factors should be easily obtainable, inexpensive, and avoid invasive clinical procedures. The aim of our study is to fill this knowledge gap by systematically mapping all the available evidence on the association of various clinical, demographic, and lifestyle variables with the risk of specific adverse outcomes in patients with COVID-19. METHODS: The systematic review was conducted using standardized methodology, searching two electronic databases (PubMed and SCOPUS) for relevant literature published between 1st January 2020 and 9th July 2020. Included studies reported characteristics of patients with COVID-19 while reporting outcomes relating to disease severity. In the case of sufficient comparable data, meta-analyses were conducted to estimate risk of each variable. RESULTS: Seventy-six studies were identified, with a total of 17,860,001 patients across 14 countries. The studies were highly heterogeneous in terms of the sample under study, outcomes, and risk measures reported. A large number of risk factors were presented for COVID-19. Commonly reported variables for adverse outcome from COVID-19 comprised patient characteristics, including age >75 (OR: 2.65, 95% CI: 1.81-3.90), male sex (OR: 2.05, 95% CI: 1.39-3.04) and severe obesity (OR: 2.57, 95% CI: 1.31-5.05). Active cancer (OR: 1.46, 95% CI: 1.04-2.04) was associated with increased risk of severe outcome. A number of common symptoms and vital measures (respiratory rate and SpO2) also suggested elevated risk profiles. CONCLUSIONS: Based on the findings of this study, a range of easily assessed parameters are valuable to predict elevated risk of severe illness and mortality as a result of COVID-19, including patient characteristics and detailed comorbidities, alongside the novel inclusion of real-time symptoms and vital measurements. DOI: 10.1371/journal.pone.0247461 PMCID: PMC7932512 PMID: 33661992 [Indexed for MEDLINE] Conflict of interest statement: A.B, A.B.R., S.P., D.P., A.Y., M.A., are employees of Huma Therapeutics Ltd. D.M & AL declare that they have no conflict of interests to report. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
http://www.ncbi.nlm.nih.gov/pubmed/32949592
1. Metabolism. 2021 Apr;117:154373. doi: 10.1016/j.metabol.2020.154373. Epub 2020 Sep 16. Association of body mass index (BMI) with critical COVID-19 and in-hospital mortality: A dose-response meta-analysis. Du Y(1), Lv Y(2), Zha W(3), Zhou N(4), Hong X(5). Author information: (1)MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha, Hunan Province 410081, China. (2)Key Laboratory of Molecular Epidemiology, Medical College of Hunan Normal University, Changsha, Hunan Province 410013, China. Electronic address: [email protected]. (3)Key Laboratory of Molecular Epidemiology, Medical College of Hunan Normal University, Changsha, Hunan Province 410013, China. Electronic address: [email protected]. (4)Key Laboratory of Molecular Epidemiology, Medical College of Hunan Normal University, Changsha, Hunan Province 410013, China. (5)Hunan Provincial People's Hospital, Scientific Research, Department, Changsha, Hunan Province 410005, China. BACKGROUND AND PURPOSE: The coronavirus disease 2019 (COVID-19) pandemic presents an unprecedented health crisis to the entire world. As reported, the body mass index (BMI) may play an important role in COVID-19; however, this still remains unclear. The aim of this study was to explore the association between BMI and COVID-19 severity and mortality. METHODS: The Medline, PubMed, Embase and Web of science were systematically searched until August 2020. Random-effects models and dose-response meta-analysis were used to synthesize the results. Combined odds ratios (ORs) with their 95% confidence intervals (CIs) were calculated, and the effect of covariates were analyzed using subgroup analysis and meta-regression analyses. RESULTS: A total of 16 observational studies involving 109,881 patients with COVID-19 were included in the meta-analysis. The pooled results showed that patients with a BMI ≥ 30 kg/m2 had a 2.35-fold risk (OR = 2.35, 95%CI = 1.64-3.38, P < 0.001) for critical COVID-19 and a 2.68-fold risk for COVID-19 mortality (OR = 2.68, 95%CI = 1.65-4.37, P < 0.001) compared with patients with a BMI <30 kg/m2. Subgroup analysis results showed that patients with obesity and age > 60 years was associated with a significantly increased risk of critical COVID-19 (OR = 3.11, 95%CI = 1.73-5.61, P < 0.001) and COVID-19 mortality (OR = 3.93, 95%CI = 2.18-7.09, P < 0.001). Meta-regression analysis results also showed that age had a significant influence on the association between BMI and COVID-19 mortality (Coef. = 0.036, P = 0.048). Random-effects dose-response meta-analysis showed a linear association between BMI and both critical COVID-19(Pnon-linearity = 0.242) and mortality (Pnon-linearity = 0.116). The risk of critical COVID-19 and mortality increased by 9%(OR = 1.09, 95%CI = 1.04-1.14, P < 0.001) and 6%(OR = 1.06, 95%CI = 1.02-1.10, P = 0.002) for each 1 kg/m2 increase in BMI, respectively. CONCLUSIONS: Evidence from this meta-analysis suggested that a linear dose-response association between BMI and both COVID-19 severity and mortality. Further, obesity (BMI ≥ 30 kg/m2) was associated with a significantly increased risk of critical COVID-19 and in-hospital mortality of COVID-19. Copyright © 2020. Published by Elsevier Inc. DOI: 10.1016/j.metabol.2020.154373 PMCID: PMC7493748 PMID: 32949592 [Indexed for MEDLINE] Conflict of interest statement: Declaration of competing interest The authors declare that there are no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32491116
1. Rev Saude Publica. 2020;54:60. doi: 10.11606/s1518-8787.2020054002481. Epub 2020 Jun 1. A brief-review of the risk factors for covid-19 severity. Rod JE(1), Oviedo-Trespalacios O(2), Cortes-Ramirez J(1). Author information: (1)School of Public Health and Social Work, Queensland University of Technology, Brisbane, Queensland, Australia. (2)Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia. The World Health Organization has emphasized that one of the most important questions to address regarding the covid-19 pandemic is to understand risk factors for disease severity. We conducted a brief review that synthesizes the available evidence and provides a judgment on the consistency of the association between risk factors and a composite end-point of severe-fatal covid-19. Additionally, we also conducted a comparability analysis of risk factors across 17 studies. We found evidence supporting a total of 60 predictors for disease severity, of which seven were deemed of high consistency, 40 of medium and 13 of low. Among the factors with high consistency of association, we found age, C-reactive protein, D-dimer, albumin, body temperature, SOFA score and diabetes. The results suggest that diabetes might be the most consistent comorbidity predicting disease severity and that future research should carefully consider the comparability of reporting cases, factors, and outcomes along the different stages of the natural history of covid-19. DOI: 10.11606/s1518-8787.2020054002481 PMCID: PMC7263798 PMID: 32491116 [Indexed for MEDLINE] Conflict of interest statement: Conflict of Interest: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32472676
1. Clin Infect Dis. 2020 Nov 5;71(8):1962-1968. doi: 10.1093/cid/ciaa674. Predictors for Severe COVID-19 Infection. Bhargava A(1), Fukushima EA(1), Levine M(1), Zhao W(1), Tanveer F(1), Szpunar SM(1), Saravolatz L(1). Author information: (1)Ascension St John Hospital, Detroit, Michigan, USA. Comment in Clin Infect Dis. 2021 May 18;72(10):1868-1869. doi: 10.1093/cid/ciaa956. Clin Infect Dis. 2021 May 18;72(10):1870. doi: 10.1093/cid/ciaa960. BACKGROUND: COVID-19 is a pandemic disease caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Predictors for severe COVID-19 infection have not been well defined. Determination of risk factors for severe infection would enable identifying patients who may benefit from aggressive supportive care and early intervention. METHODS: We conducted a retrospective observational study of 197 patients with confirmed COVID-19 admitted to a tertiary academic medical center. RESULTS: Of 197 hospitalized patients, the mean (SD) age of the cohort was 60.6 (16.2) years, 103 (52.3%) were male, and 156 (82.1%) were black. Severe COVID-19 infection was noted in 74 (37.6%) patients, requiring intubation. Patients aged above 60 were significantly more likely to have severe infection. Patients with severe infection were significantly more likely to have diabetes, renal disease, and chronic pulmonary disease and had significantly higher white blood cell counts, lower lymphocyte counts, and increased C-reactive protein (CRP) than patients with nonsevere infection. In multivariable logistic regression analysis, risk factors for severe infection included pre-existing renal disease (odds ratio [OR], 7.4; 95% CI, 2.5-22.0), oxygen requirement at hospitalization (OR, 2.9; 95% CI, 1.3-6.7), acute renal injury (OR, 2.7; 95% CI, 1.3-5.6), and CRP on admission (OR, 1.006; 95% CI, 1.001-1.01). Race, age, and socioeconomic status were not independent predictors. CONCLUSIONS: Acute or pre-existing renal disease, supplemental oxygen upon hospitalization, and admission CRP were independent predictors for the development of severe COVID-19. Every 1-unit increase in CRP increased the risk of severe disease by 0.06%. © The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: [email protected]. DOI: 10.1093/cid/ciaa674 PMCID: PMC7314166 PMID: 32472676 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35042956
1. Pediatr Res. 2022 Oct;92(4):1140-1145. doi: 10.1038/s41390-021-01902-y. Epub 2022 Jan 19. No infectious SARS-CoV-2 in breast milk from a cohort of 110 lactating women. Krogstad P(1)(2), Contreras D(3), Ng H(3), Tobin N(3), Chambers CD(4)(5), Bertrand K(4)(5), Bode L(4)(6), Aldrovandi GM(3). Author information: (1)Department of Pediatrics, David Geffen Scool of Medicine at UCLAs, University of California, Los Angeles, CA, USA. [email protected]. (2)Departments of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA. [email protected]. (3)Department of Pediatrics, David Geffen Scool of Medicine at UCLAs, University of California, Los Angeles, CA, USA. (4)Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA. (5)Hebert Wertheim School of Public Health and Longevity Science, University of California, San Diego, La Jolla, CA, USA. (6)Larsson-RosenquistFoundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, USA. Update of medRxiv. 2021 Apr 07:2021.04.05.21254897. doi: 10.1101/2021.04.05.21254897. BACKGROUND: Genomic RNA of severe acute respiratory syndrome-associated coronavirus type 2 (SARS-CoV-2) has been detected in the breast milk of lactating women, but its pathological significance has remained uncertain due to the small size of prior studies. METHODS: Breast milk from 110 lactating women was analyzed by reverse transcription-polymerase chain reaction (285 samples) and viral culture (160 samples). Those containing SARS-CoV-2 viral RNA (vRNA) were examined for the presence of subgenomic RNA (sgRNA), a putative marker of infectivity. RESULTS: Sixty-five women had a positive SARS-CoV-2 diagnostic test, 9 had symptoms but negative diagnostic tests, and 36 symptomatic women were not tested. SARS-CoV-2 vRNA was detected in the milk of 7 (6%) women with either a confirmed infection or symptomatic illness, including 6 of 65 (9%) women with a positive SARS-CoV-2 diagnostic test. Infectious virus was not detected in any culture and none had detectable sgRNA. In control experiments, infectious SARS-CoV-2 could be cultured after addition to breastmilk despite several freeze-thaw cycles, as it occurs in the storage and usage of human milk. CONCLUSIONS: SARS-CoV-2 RNA can be found infrequently in the breastmilk after recent infection, but we found no evidence that breastmilk contains an infectious virus or that breastfeeding represents a risk factor for transmission of infection to infants. IMPACT: This article goes beyond prior small studies to provide evidence that infectious SARS-CoV-2 is not present in the milk of lactating women with recent infection, even when SARS-CoV-2 RNA is detected. Recent SARS-CoV-2 infection or detection of its RNA in human milk is not a contraindication to breastfeeding. © 2021. The Author(s). DOI: 10.1038/s41390-021-01902-y PMCID: PMC9586866 PMID: 35042956 [Indexed for MEDLINE] Conflict of interest statement: L.B. reported serving as the UC San Diego Chair of Collaborative Human Milk Research, which is endowed by the Family Larsson-Rosenquist Foundation. Medela Corporation provided milk sample collection materials for this study. C.D.C. reports that shipping of milk samples was financially supported by the Mothers’ Milk Bank at Austin, an accredited milk bank and member of the Human Milk Banking Association of North America.
http://www.ncbi.nlm.nih.gov/pubmed/33555566
1. Indian J Pediatr. 2021 Aug;88(8):800-801. doi: 10.1007/s12098-021-03681-0. Epub 2021 Feb 8. Is SARS-CoV-2 Transmitted Through Breastfeeding? Thanigainathan S(1), Kaliyaperumal V(2), Sivanandan S(1), Rengaraj S(3), Dhodapkar R(2), Bethou A(4). Author information: (1)Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India. (2)Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India. (3)Department of Obstetrics and Gynecology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, India. (4)Department of Neonatology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India. [email protected]. There are concerns regarding the transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from mother to child during this COVID pandemic. This descriptive study was done to check the possible transmission of the virus through breastfeeding in the Indian context. RT-qPCR for SARS-CoV-2 was done in breast milk samples from 30 COVID-positive mothers. Paired oropharyngeal swabs of the same neonates were also sent for RT-PCR at 48 h and on day 5 of life. All the breast milk samples were negative for SARS-CoV-2 except one. A repeat sample of breast milk from the same mother was also negative when rechecked the next day. All the paired neonatal oropharyngeal swabs were also negative for SARS-CoV-2. The authors could not find evidence for transmission of SARS-CoV-2 from mother to child through breastmilk in the population studied. © 2021. Dr. K C Chaudhuri Foundation. DOI: 10.1007/s12098-021-03681-0 PMCID: PMC7868520 PMID: 33555566 [Indexed for MEDLINE] Conflict of interest statement: None.
http://www.ncbi.nlm.nih.gov/pubmed/32995804
1. medRxiv [Preprint]. 2020 Sep 18:2020.09.16.20196071. doi: 10.1101/2020.09.16.20196071. COVID-19 and human milk: SARS-CoV-2, antibodies, and neutralizing capacity. Pace RM(1), Williams JE(2), Järvinen KM(3), Belfort MB(4), Pace CDW(1), Lackey KA(1), Gogel AC(1), Nguyen-Contant P(5), Kanagaiah P(5), Fitzgerald T(5), Ferri R(3), Young B(3), Rosen-Carole C(3), Diaz N(3), Meehan CL(6), Caffe B(6), Sangster MY(5), Topham D(5), McGuire MA(2), Seppo A(3), McGuire MK(1). Author information: (1)Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID 83844. (2)Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID 83844. (3)Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642. (4)Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115. (5)David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642. (6)Department of Anthropology, Washington State University, Pullman, WA 99164. Update in This article has been published with doi: 10.1128/mbio.03192-20. BACKGROUND: It is not known whether SARS-CoV-2 can be transmitted from mother to infant during breastfeeding, and if so whether the benefits of breastfeeding outweigh this risk. This study was designed to evaluate 1) if SARS-CoV-2 RNA can be detected in milk and on the breast of infected women, 2) concentrations of milk-borne anti-SARS-CoV-2 antibodies, and 3) the capacity of milk to neutralize SARS-CoV-2 infectivity. METHODS: We collected 37 milk samples and 70 breast swabs (before and after breast washing) from 18 women recently diagnosed with COVID-19. Samples were analyzed for SARS-CoV-2 RNA using RT-qPCR. Milk was also analyzed for IgA and IgG specific for the nucleocapsid protein, receptor binding domain (RBD), S2 subunit of the spike protein of SARS-CoV-2, as well as 2 seasonal coronaviruses using ELISA; and for its ability to neutralize SARS-CoV-2. RESULTS: We did not detect SARS-CoV-2 RNA in any milk sample. In contrast, SARS-CoV-2 RNA was detected on several breast swabs, although only one was considered conclusive. All milk contained SARS-CoV-2-specific IgA and IgG, and levels of anti-RBD IgA correlated with SARS-CoV-2 neutralization. Strong correlations between levels of IgA and IgG to SARS-CoV-2 and seasonal coronaviruses were noted. CONCLUSIONS: Our data do not support maternal-to-child transmission of SARS-CoV-2 via milk; however, risk of transmission via breast skin should be further evaluated. Importantly, milk produced by infected mothers is a source of anti-SARS-CoV-2 IgA and IgG and neutralizes SARS-CoV-2 activity. These results support recommendations to continue breastfeeding during mild-to-moderate maternal COVID-19 illness. DOI: 10.1101/2020.09.16.20196071 PMCID: PMC7523143 PMID: 32995804
http://www.ncbi.nlm.nih.gov/pubmed/33394459
1. Curr Nutr Rep. 2021 Mar;10(1):71-75. doi: 10.1007/s13668-020-00343-z. Epub 2021 Jan 4. Should COVID-19 Mother Breastfeed her Newborn Child? A Literature Review on the Safety of Breastfeeding for Pregnant Women with COVID-19. Bhatt H(1)(2). Author information: (1)Goshen Hospital, Goshen, IN, USA. [email protected]. (2)Indiana University School of Medicine, South Bend, IN, USA. [email protected]. PURPOSE OF REVIEW: Breastfeeding is beneficial to both the newborn and the mother. During the COVID-19 pandemic, concerns have been raised on whether the SARS-CoV-2 virus could be transmitted from COVID-19 positive mother to the newborn through breastmilk. The purpose of this review is to examine the available evidence on the risks of transmission of infection from COVID-19 mothers to their newborns through breastfeeding. RECENT FINDINGS: Data is very limited in this regard, with only a few smaller case series, and case reports have been published so far. In most of the studies, breastmilk samples from COVID-19 mothers tested negative for the virus. In the case reports where the virus was detected in breastmilk and the infants were diagnosed with COVID-19, it remained unclear whether the disease was transmitted through breastmilk or direct contact or through delivery. Another hypothesis is that the viral antibodies could pass to the newborn passively through breastmilk of COVID-19 positive mothers and give immunity to the child, but data is minimal. Based on the currently available limited evidence and recognizing the benefits of breastfeeding, it may be concluded that if the health of the mother and her newborn allows, direct breastfeeding or extracted breastmilk should be encouraged by the healthcare providers after a careful discussion of the risks of vertical transmission to the mother and her family. Preventive measures should be taken by COVID-19 mothers to prevent droplet transmission of infection to the infants while breastfeeding. DOI: 10.1007/s13668-020-00343-z PMCID: PMC7780073 PMID: 33394459 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32860259
1. Ann N Y Acad Sci. 2021 Jan;1484(1):32-54. doi: 10.1111/nyas.14477. Epub 2020 Aug 28. Transmission of SARS-CoV-2 through breast milk and breastfeeding: a living systematic review. Centeno-Tablante E(1), Medina-Rivera M(1), Finkelstein JL(1), Rayco-Solon P(2), Garcia-Casal MN(3), Rogers L(3), Ghezzi-Kopel K(4), Ridwan P(1), Peña-Rosas JP(3), Mehta S(1). Author information: (1)Division of Nutritional Sciences, Cornell University, Ithaca, New York. (2)Department of Maternal, Newborn, Child and Adolescent Health and Ageing, World Health Organization, Geneva, Switzerland. (3)Department of Nutrition and Food Safety, World Health Organization, Geneva, Switzerland. (4)Albert R. Mann Library, Cornell University, Ithaca, New York. The pandemic of coronavirus disease 2019 (COVID-19) is caused by infection with a novel coronavirus strain, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). At present, there is limited information on potential transmission of the infection from mother to child, particularly through breast milk and breastfeeding. Here, we provide a living systematic review to capture information that might necessitate changes in the guidance on breast milk and breastfeeding given the uncertainty in this area. Our search retrieved 19,414 total records; 605 were considered for full-text eligibility and no ongoing trials were identified. Our review includes 340 records, 37 with breast milk samples and 303 without. The 37 articles with analyzed breast milk samples reported on 77 mothers who were breastfeeding their children; among them, 19 of 77 children were confirmed COVID-19 cases based on RT-PCR assays, including 14 neonates and five older infants. Nine of the 68 analyzed breast milk samples from mothers with COVID-19 were positive for SARS-CoV-2 RNA; of the exposed infants, four were positive and two were negative for COVID-19. Currently, there is no evidence of SARS-CoV-2 transmission through breast milk. Studies are needed with longer follow-up periods that collect data on infant feeding practices and on viral presence in breast milk. © 2020 The Authors. Annals of the New York Academy of Sciences. The World Health Organization retains copyright and all other rights in the manuscript of this article as submitted for publication. DOI: 10.1111/nyas.14477 PMCID: PMC7970667 PMID: 32860259 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33344466
1. Front Med (Lausanne). 2020 Dec 2;7:562700. doi: 10.3389/fmed.2020.562700. eCollection 2020. Persistent SARS-CoV-2 RNA Positive in Feces but Negative in Breastmilk: A Case Report of COVID-19 in a Breastfeeding Patient. Chu H(1), Li J(1), Yan J(1), Bai T(1), Schnabl B(2), Zou L(3), Yang L(1), Hou X(1). Author information: (1)Division of Gastroenterology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China. (2)Department of Medicine, University of California, San Diego, La Jolla, CA, United States. (3)Department of Obstetrics & Gynecology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China. COVID-19 is a pandemic infectious disease. Whether SARS-CoV-2 was transmitted through breast milk is unknown. Here, we report a breastfeeding woman with COVID-19 presenting with gastrointestinal symptoms and persistent SARS-CoV-2 RNA positivity in both her oropharyngeal swabs and feces, but negativity in her breastmilk. After appearance of serum SARS-CoV-2-IgG, she began to bottle feed her baby with breastmilk without transmission. This report facilitates the understanding of breastfeeding-related risks in COVID-19. Copyright © 2020 Chu, Li, Yan, Bai, Schnabl, Zou, Yang and Hou. DOI: 10.3389/fmed.2020.562700 PMCID: PMC7738631 PMID: 33344466 Conflict of interest statement: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/33538539
1. Pediatr Infect Dis J. 2021 Apr 1;40(4):e137-e145. doi: 10.1097/INF.0000000000003043. Risk Factors for Severe COVID-19 in Children. Graff K(1), Smith C(1), Silveira L(1), Jung S(2), Curran-Hays S(1), Jarjour J(1), Carpenter L(3), Pickard K(3), Mattiucci M(1), Fresia J(3), McFarland EJ(1), Dominguez SR(1)(2), Abuogi L(1). Author information: (1)Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO. (2)Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, Aurora, CO. (3)University of Colorado School of Medicine, Aurora, CO. BACKGROUND: There are limited pediatric data regarding severe COVID-19 disease. Our study aims to describe the epidemiology and identify risk factors for severe COVID-19 disease in children. METHODS: This is a retrospective cohort study among children with positive SARS-CoV-2 PCR from March to July 2020 at Children's Hospital Colorado. Risk factors for severe disease were analyzed as defined by hospital admission, respiratory support, or critical care. Univariable and multivariable analyses were conducted. RESULTS: Among 454 patients identified with SARS-CoV-2, 191 (42.1%) were females, median age 11 years. Fifty-five percent of all patients identified as Hispanic compared with 29% among all hospital visits in 2019 (P < 0.0001). In multivariable analyses, age 0-3 months or >20 years [adjusted odds ratio (aOR), 7.85; P < 0.0001 and aOR, 5.1; P = 0.03, respectively], preterm birth history (aOR, 3.7; P = 0.03), comorbidities [including immunocompromise (aOR, 3.5; P = 0.004), gastrointestinal condition (aOR, 2.7; P = 0.009), diabetes (aOR, 6.6; P = 0.04), asthma (aOR, 2.2; P = 0.04)], and specific symptoms at presentation were predictors for admission. Age 0-3 months or >20 years, asthma, gastrointestinal condition, and similar symptoms at presentation were also predictors for respiratory support. Elevated C-reactive protein was associated with the need for critical care with median of 17.7 mg/dL (IQR, 5.3-22.9) versus 1.95 mg/dL (IQR, 0.7-5.5) among patients requiring critical versus no critical care (OR, 1.2; P = 0.02). CONCLUSIONS: Extremes of age, comorbid conditions, and elevated CRP are predictors of severe disease in children. Findings from this study can inform pediatric providers and public health officials to tailor clinical management, pandemic planning, and resource allocation. Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved. DOI: 10.1097/INF.0000000000003043 PMID: 33538539 [Indexed for MEDLINE] Conflict of interest statement: The authors have no conflicts of interest to disclose.
http://www.ncbi.nlm.nih.gov/pubmed/34013217
1. Lancet Reg Health West Pac. 2020 Nov;4:100045. doi: 10.1016/j.lanwpc.2020.100045. Epub 2020 Nov 10. A study of breastfeeding practices, SARS-CoV-2 and its antibodies in the breast milk of mothers confirmed with COVID-19. Peng S(1), Zhu H(1), Yang L(2), Cao L(1), Huang X(3), Dynes M(4), Narayan A(3), Xia J(5), Chen Y(1), Zhang P(1), Liu H(1), Li H(1), Xia S(1). Author information: (1)Department of Neonatology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei 430070, China. (2)Department of Neonatology, Xiaochang First People's Hospital, Xiaochang, Hubei, China. (3)Health, Nutrition and WASH, UNICEF China, Beijing, China. (4)UNICEF East Asia and Pacific Regional Office, Bangkok, Thailand. (5)Department of Laboratory Medicine, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, China. BACKGROUND: The possibility of 2019 novel coronavirus disease (COVID-19) transmission to neonates through breast milk remains unverified. METHODS: This paper presents the interim results of a longitudinal study being carried out in Hubei province. As of 1 April 2020, 24 mothers confirmed with COVID-19, 19 mothers suspected with COVID-19 but Polymerase chain reaction negative, and 21 mothers without COVID-19 and their neonates have been recruited. Telephone follow-up was conducted to collect information on breastfeeding practices. Forty-four breast milk samples were collected from 16 of the 24 mothers with confirmed COVID-19 for the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) ribonucleic acid (RNA) and antibodies (IgM and IgG) testing. FINDINGS: The average mother-child separation time was 36•7 ± 21•1 days among mothers confirmed with COVID-19, significantly longer than that of the suspected group (16•6 ± 13•1 days) and control group (10•5 ± 8•2 days). Both the COVID-19 confirmed (58•3%) and suspected (52•6%) groups presented significantly lower rates of breastfeeding as compared with the control group (95•2%). All 44 breast milk samples tested negative for the SARS-CoV-2 nucleic acid. Thirty-eight breast milk samples underwent antibody testing and all tested negative for IgG. Twenty-one breast milk samples from 8 women tested positive for IgM, while the remaining samples from 11 women tested negative. INTERPRETATION: Considering the lack of evidence for SARS-CoV-2 transmission through breast milk, breastfeeding counselling along with appropriate hand hygiene precautions and facemasks should be provided to all pregnant women. FUNDING: The study was funded by the Hong Kong Committee for UNICEF. © 2020 Published by Elsevier Ltd. DOI: 10.1016/j.lanwpc.2020.100045 PMCID: PMC7654387 PMID: 34013217 Conflict of interest statement: The authors of this paper have no conflict of interest. The opinions expressed in this paper are solely those of the authors and do not necessarily represent the official position of UNICEF.
http://www.ncbi.nlm.nih.gov/pubmed/34182576
1. Am J Perinatol. 2021 Sep;38(11):1209-1216. doi: 10.1055/s-0041-1731451. Epub 2021 Jun 28. Breast Milk and Breastfeeding of Infants Born to SARS-CoV-2 Positive Mothers: A Prospective Observational Cohort Study. Kunjumon B(1), Wachtel EV(1), Lumba R(1), Quan M(1), Remon J(1), Louie M(1), Verma S(1), Moffat MA(1), Kouba I(2), Bennett TA(3)(2), Mejia CM(4), Mally PV(1), Lin X(4), Hanna N(4). Author information: (1)Department of Pediatrics, New York University Grossman School of Medicine, New York City, New York. (2)Department of Obstetrics and Gynecology, New York University Grossman School of Medicine, New York City, New York. (3)Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, New York University Grossman School of Medicine, New York City, New York. (4)Department of Pediatrics, New York University Long Island School of Medicine, New York City, New York. OBJECTIVE: There are limited published data on the transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus from mothers to newborns through breastfeeding or from breast milk. The World Health Organization released guidelines encouraging mothers with suspected or confirmed COVID-19 to breastfeed as the benefits of breastfeeding outweighs the possible risk of transmission. The objective of this study was to determine if SARS-CoV-2 was present in the breast milk of lactating mothers who had a positive SARS-CoV-2 nasopharyngeal swab test prior to delivery, and the clinical outcomes for their newborns. STUDY DESIGN: This was a single-center, observational, prospective cohort study. Maternal-newborn dyads that delivered at New York University Langone Hospital Brooklyn with confirmed maternal SARS-CoV-2 positive screen test at the time of admission were recruited for the study. Breast milk samples were collected during postpartum hospitalization and tested for the presence of SARS-CoV-2 genes N1 and N2 by two-step reverse transcription polymerase chain reaction. Additionally, the clinical characteristics of the maternal newborn dyad, results of nasopharyngeal SARS-CoV-2 testing, and neonatal follow-up data were collected. RESULTS: A total of 19 mothers were included in the study and their infants who were all fed breast milk. Breast milk samples from 18 mothers tested negative for SARS-CoV-2, and 1 was positive for SARS-CoV-2 RNA. The infant who ingested the breast milk that tested positive had a negative nasopharyngeal test for SARS-CoV-2, and had a benign clinical course. There was no evidence of significant clinical infection during the hospital stay or from outpatient neonatal follow-up data for all the infants included in this study. CONCLUSION: In a small cohort of SARS-CoV-2 positive lactating mothers giving birth at our institution, most of their breast milk samples (95%) contained no detectable virus, and there was no evidence of COVID-19 infection in their breast milk-fed neonates. KEY POINTS: · Breast milk may rarely contain detectable SARS-CoV-2 RNA and was not detected in asymptomatic mothers.. · Breast milk with detectable SARS-CoV-2 RNA from a symptomatic mother had no clinical significance for her infant.. · Breast feeding with appropriate infection control instructions appears to be safe in mother with COVID infection.. Thieme. All rights reserved. DOI: 10.1055/s-0041-1731451 PMID: 34182576 [Indexed for MEDLINE] Conflict of interest statement: None declared.
http://www.ncbi.nlm.nih.gov/pubmed/32472745
1. Matern Child Nutr. 2020 Oct;16(4):e13032. doi: 10.1111/mcn.13032. Epub 2020 May 30. SARS-CoV-2 and human milk: What is the evidence? Lackey KA(1), Pace RM(1), Williams JE(2), Bode L(3), Donovan SM(4), Järvinen KM(5), Seppo AE(5), Raiten DJ(6), Meehan CL(7), McGuire MA(2), McGuire MK(1). Author information: (1)Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho, USA. (2)Department of Animal and Veterinary Sciences, University of Idaho, Moscow, Idaho, USA. (3)Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, California, USA. (4)Department of Food Science and Human Nutrition and Institute of Genomic Biology, University of Illinois, Urbana, Illinois, USA. (5)Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA. (6)Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA. (7)Department of Anthropology, Washington State University, Pullman, Washington, USA. Update of medRxiv. 2020 Apr 11:2020.04.07.20056812. doi: 10.1101/2020.04.07.20056812. The novel coronavirus SARS-CoV-2 has emerged as one of the most compelling and concerning public health challenges of our time. To address the myriad issues generated by this pandemic, an interdisciplinary breadth of research, clinical and public health communities has rapidly engaged to collectively find answers and solutions. One area of active inquiry is understanding the mode(s) of SARS-CoV-2 transmission. Although respiratory droplets are a known mechanism of transmission, other mechanisms are likely. Of particular importance to global health is the possibility of vertical transmission from infected mothers to infants through breastfeeding or consumption of human milk. However, there is limited published literature related to vertical transmission of any human coronaviruses (including SARS-CoV-2) via human milk and/or breastfeeding. Results of the literature search reported here (finalized on 17 April 2020) revealed a single study providing some evidence of vertical transmission of human coronavirus 229E; a single study evaluating presence of SARS-CoV in human milk (it was negative); and no published data on MERS-CoV and human milk. We identified 13 studies reporting human milk tested for SARS-CoV-2; one study (a non-peer-reviewed preprint) detected the virus in one milk sample, and another study detected SARS-CoV-2 specific IgG in milk. Importantly, none of the studies on coronaviruses and human milk report validation of their collection and analytical methods for use in human milk. These reports are evaluated here, and their implications related to the possibility of vertical transmission of coronaviruses (in particular, SARS-CoV-2) during breastfeeding are discussed. © 2020 The Authors. Maternal & Child Nutrition published by John Wiley & Sons Ltd. DOI: 10.1111/mcn.13032 PMCID: PMC7300480 PMID: 32472745 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/33563823
1. mBio. 2021 Feb 9;12(1):e03192-20. doi: 10.1128/mBio.03192-20. Characterization of SARS-CoV-2 RNA, Antibodies, and Neutralizing Capacity in Milk Produced by Women with COVID-19. Pace RM(#)(1), Williams JE(#)(2), Järvinen KM(3), Belfort MB(4), Pace CDW(1), Lackey KA(1), Gogel AC(1), Nguyen-Contant P(5), Kanagaiah P(5), Fitzgerald T(5), Ferri R(3), Young B(3), Rosen-Carole C(3), Diaz N(3), Meehan CL(6), Caffé B(6), Sangster MY(5), Topham D(5), McGuire MA(2), Seppo A(7), McGuire MK(8). Author information: (1)Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho, USA. (2)Department of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, Idaho, USA. (3)Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA. (4)Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA. (5)David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA. (6)Department of Anthropology, Washington State University, Pullman, Washington, USA. (7)Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA [email protected] [email protected]. (8)Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho, USA [email protected] [email protected]. (#)Contributed equally Whether mother-to-infant SARS-CoV-2 transmission can occur during breastfeeding and, if so, whether the benefits of breastfeeding outweigh this risk during maternal COVID-19 illness remain important questions. Using RT-qPCR, we did not detect SARS-CoV-2 RNA in any milk sample (n = 37) collected from 18 women following COVID-19 diagnosis. Although we detected evidence of viral RNA on 8 out of 70 breast skin swabs, only one was considered a conclusive positive result. In contrast, 76% of the milk samples collected from women with COVID-19 contained SARS-CoV-2-specific IgA, and 80% had SARS-CoV-2-specific IgG. In addition, 62% of the milk samples were able to neutralize SARS-CoV-2 infectivity in vitro, whereas milk samples collected prior to the COVID-19 pandemic were unable to do so. Taken together, our data do not support mother-to-infant transmission of SARS-CoV-2 via milk. Importantly, milk produced by infected mothers is a beneficial source of anti-SARS-CoV-2 IgA and IgG and neutralizes SARS-CoV-2 activity. These results support recommendations to continue breastfeeding during mild-to-moderate maternal COVID-19 illness.IMPORTANCE Results from prior studies assaying human milk for the presence of SARS-CoV-2, the causative virus of COVID-19, have suggested milk may act as a potential vehicle for mother-to-child transmission. Most previous studies are limited because they followed only a few participants, were cross-sectional, and/or failed to report how milk was collected and/or analyzed. As such, considerable uncertainty remains regarding whether human milk is capable of transmitting SARS-CoV-2 from mother to child. Here, we report that repeated milk samples collected from 18 women following COVID-19 diagnosis did not contain SARS-CoV-2 RNA; however, risk of transmission via breast skin should be further evaluated. Importantly, we found that milk produced by infected mothers is a source of anti-SARS-CoV-2 IgA and IgG and neutralizes SARS-CoV-2 activity. These results support recommendations to continue breastfeeding during mild-to-moderate maternal COVID-19 illness as milk likely provides specific immunologic benefits to infants. Copyright © 2021 Pace et al. DOI: 10.1128/mBio.03192-20 PMCID: PMC7885115 PMID: 33563823 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32454808
1. Rev Panam Salud Publica. 2020 Apr 27;44:e59. doi: 10.26633/RPSP.2020.59. eCollection 2020. To breastfeed or not to breastfeed? Lack of evidence on the presence of SARS-CoV-2 in breastmilk of pregnant women with COVID-19. Martins-Filho PR(1), Santos VS(2), Santos HP Jr(3). Author information: (1)Federal University of Sergipe Federal University of Sergipe São Cristóvão Brazil Federal University of Sergipe, São Cristóvão, Brazil. (2)Universidade Federal de Alagoas Universidade Federal de Alagoas Arapiraca Brazil Universidade Federal de Alagoas, Arapiraca, Brazil. (3)University of North Carolina at Chapel Hill University of North Carolina at Chapel Hill Chapel Hill United States of America University of North Carolina at Chapel Hill, Chapel Hill, United States of America. A rapid systematic review was carried out to evaluate the current evidence related to the presence of SARS-CoV-2 in breast milk from pregnant women with COVID-19. Eight studies analyzing the presence of SARS-CoV-2 RNA in the breast milk of 24 pregnant women with COVID-19 during the third trimester of pregnancy were found. All patients had fever and/or symptoms of acute respiratory illness and chest computed tomography images indicative of COVID-19 pneumonia. Most pregnant women had cesarean delivery (91.7%) and two neonates had low birthweight (< 2 500 g). Biological samples collected immediately after birth from upper respiratory tract (throat or nasopharyngeal) of neonates and placental tissues showed negative results for the presence SARS-CoV-2 by RT-PCR test. No breast milk samples were positive for SARS-CoV-2 and, to date, there is no evidence on the presence of SARS-CoV-2 in breast milk of pregnant women with COVID-19. However, data are still limited and breastfeeding of women with COVID-19 remains a controversial issue. There are no restrictions on the use of milk from a human breast milk bank. Publisher: Se llevó a cabo una revisión sistemática rápida para evaluar la evidencia disponible sobre la presencia de SARS-CoV-2 en la leche materna de mujeres embarazadas afectadas con COVID-19. Se encontraron ocho estudios que analizaron la presencia de ARN de SARS-CoV-2 en la leche materna de 24 mujeres embarazadas con COVID-19 durante el tercer trimestre del embarazo. Todas las pacientes tenían fiebre o síntomas de enfermedad respiratoria aguda e imágenes de tomografía computarizada de tórax indicativas de neumonía por COVID-19. La mayoría de las mujeres embarazadas (91,7%) tuvieron un parto por cesárea y dos neonatos presentaron bajo peso al nacer (< 2 500 g). Las muestras biológicas recogidas inmediatamente después del parto de las vías respiratorias superiores (faringe o nasofaringe) de los neonatos y los tejidos placentarios mostraron resultados negativos para SARS-CoV-2 mediante RT-PCR. Ninguna muestra de leche materna fue positiva para SARS-CoV-2 y, hasta la fecha, no hay evidencia de la presencia de SARS-CoV-2 en la leche materna de las mujeres embarazadas con COVID-19. Sin embargo, los datos disponibles todavía son limitados y la lactancia materna en las mujeres con COVID-19 sigue siendo un tema controvertido. No hay restricciones para el uso de leche materna de banco. Publisher: Foi realizada uma revisão sistemática rápida para avaliar as evidências atuais relacionadas à presença da SARS-CoV-2 no leite materno de mulheres grávidas com COVID-19. Foram encontrados oito estudos analisando a presença de RNA do SARS-CoV-2 no leite materno de 24 gestantes com COVID-19 durante o terceiro trimestre de gravidez. Todas as pacientes apresentavam febre ou sintomas de doença respiratória aguda e imagens de tomografia computadorizada do tórax indicativas de pneumonia pela COVID-19. A maioria das gestantes teve parto cesáreo (91,7%) e dois recém-nascidos tiveram baixo peso ao nascer (< 2 500 g). As amostras biológicas coletadas imediatamente após o nascimento do trato respiratório superior (faringe ou nasofaringe) de neonatos e tecidos placentários apresentaram resultados negativos para a presença do SARS-CoV-2 pelo teste RT-PCR. Nenhuma amostra de leite materno foi positiva para o SARS-CoV-2 e, até à data, não há evidências da presença do SARS-CoV-2 no leite materno de mulheres grávidas com COVID-19. No entanto, os dados ainda são limitados e a amamentação de mulheres com COVID-19 continua a ser uma questão controversa. Não há restrições ao uso de leite de um banco de leite materno humano. DOI: 10.26633/RPSP.2020.59 PMCID: PMC7241574 PMID: 32454808 Conflict of interest statement: Conflict of interest. None declared.
http://www.ncbi.nlm.nih.gov/pubmed/32511431
1. medRxiv [Preprint]. 2020 Apr 11:2020.04.07.20056812. doi: 10.1101/2020.04.07.20056812. SARS-CoV-2 and human milk: what is the evidence? Lackey KA(1), Pace RM(1), Williams JE(2), Bode L(3), Donovan SM(4), Järvinen KM(5), Seppo AE(5), Raiten DJ(6), Meehan CL(7), McGuire MA(2), McGuire MK(1). Author information: (1)Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, USA. (2)Department of Animal and Veterinary Sciences, University of Idaho, Moscow, ID, USA. (3)Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA, USA. (4)Department of Food Science and Human Nutrition and Institute of Genomic Biology, University of Illinois, Urbana, IL USA. (5)Department of Pediatrics, Division of Allergy and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA. (6)Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA. (7)Department of Anthropology, Washington State University, Pullman, WA, USA. Update in Matern Child Nutr. 2020 Oct;16(4):e13032. doi: 10.1111/mcn.13032. The novel coronavirus SARS-CoV-2 has emerged as one of the most compelling public health challenges of our time. To address the myriad issues generated by this pandemic, an interdisciplinary breadth of research, clinical, and public health communities have rapidly engaged to find answers and solutions. One area of active inquiry is understanding the mode(s) of SARS-CoV-2 transmission. While respiratory droplets are a known mechanism of transmission, other mechanisms are possible. Of particular importance to global health is the possibility of vertical transmission from infected mothers to infants through breastfeeding or consumption of human milk. However, there is limited published literature related to vertical transmission of any human coronavirus (including SARS-CoV-2) via human milk and/or breastfeeding. There is a single study providing some evidence of vertical transmission of human coronavirus 229E, a single study evaluating presence of SARS-CoV in human milk (it was negative), and no published data on MERS-CoV and human milk. There are 9 case studies of human milk tested for SARS-CoV-2; none detected the virus. Importantly, none of the published studies on coronaviruses and human milk report validation of their analytical methods for use in human milk. These reports are evaluated here, and their implications related to the possibility of vertical transmission of coronaviruses (in particular, SARS-CoV-2) during breastfeeding are discussed. DOI: 10.1101/2020.04.07.20056812 PMCID: PMC7217082 PMID: 32511431 Conflict of interest statement: CONFLICT OF INTEREST STATEMENTS The authors declare no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/20444609
1. Trends Biochem Sci. 2010 Sep;35(9):476-89. doi: 10.1016/j.tibs.2010.04.001. Epub 2010 May 3. The histone shuffle: histone chaperones in an energetic dance. Das C(1), Tyler JK, Churchill ME. Author information: (1)Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA. Our genetic information is tightly packaged into a rather ingenious nucleoprotein complex called chromatin in a manner that enables it to be rapidly accessed during genomic processes. Formation of the nucleosome, which is the fundamental unit of chromatin, occurs via a stepwise process that is reversed to enable the disassembly of nucleosomes. Histone chaperone proteins have prominent roles in facilitating these processes as well as in replacing old histones with new canonical histones or histone variants during the process of histone exchange. Recent structural, biophysical and biochemical studies have begun to shed light on the molecular mechanisms whereby histone chaperones promote chromatin assembly, disassembly and histone exchange to facilitate DNA replication, repair and transcription. Copyright (c) 2010 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.tibs.2010.04.001 PMCID: PMC4004086 PMID: 20444609 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/15868422
1. Chromosome Res. 2005;13(3):289-98. doi: 10.1007/s10577-005-2166-z. Whole-genome views of chromatin structure. Lodén M(1), van Steensel B. Author information: (1)Netherlands Cancer Institute, Amsterdam, 1066CX, The Netherlands. DNA in eukaryotes is packed into chromatin. The basic component of chromatin is the nucleosome consisting of DNA wrapped around a histone octamer. Inside the cell nucleus, chromatin is folded into higher-order structures through various mechanisms, including repositioning of nucleosomes along the DNA, packing of nucleosomes into more condensed 3-dimensional configurations, looping of chromatin fibres, and tethering of chromosomal regions to nuclear structures. Over the past few years, new microarray-based methods have been developed for the genome-wide mapping of various aspects of chromatin structure. These methods are beginning to provide insights into the different types of chromatin and the architectural principles that govern the 3-dimensional organisation of the genome inside the nucleus. DOI: 10.1007/s10577-005-2166-z PMID: 15868422 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28058066
1. Biophys Rev. 2016;8(Suppl 1):33-49. doi: 10.1007/s12551-016-0212-z. Epub 2016 Oct 18. Recent insights from in vitro single-molecule studies into nucleosome structure and dynamics. Ordu O(1), Lusser A(2), Dekker NH(1). Author information: (1)Bionanoscience Department, Kavli Institute of Nanoscience,, Delft University of Technology, Van der Maasweg 9,, 2629 HZ Delft, The Netherlands. (2)Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria. Eukaryotic DNA is tightly packed into a hierarchically ordered structure called chromatin in order to fit into the micron-scaled nucleus. The basic unit of chromatin is the nucleosome, which consists of a short piece of DNA wrapped around a core of eight histone proteins. In addition to their role in packaging DNA, nucleosomes impact the regulation of essential nuclear processes such as replication, transcription, and repair by controlling the accessibility of DNA. Thus, knowledge of this fundamental DNA-protein complex is crucial for understanding the mechanisms of gene control. While structural and biochemical studies over the past few decades have provided key insights into both the molecular composition and functional aspects of nucleosomes, these approaches necessarily average over large populations and times. In contrast, single-molecule methods are capable of revealing features of subpopulations and dynamic changes in the structure or function of biomolecules, rendering them a powerful complementary tool for probing mechanistic aspects of DNA-protein interactions. In this review, we highlight how these single-molecule approaches have recently yielded new insights into nucleosomal and subnucleosomal structures and dynamics. DOI: 10.1007/s12551-016-0212-z PMCID: PMC5167136 PMID: 28058066 Conflict of interest statement: Compliance with ethical standards Funding Funding for this work was provided by the European Research Council (ERC) via a Consolidator Grant DynGenome (No:312221) to N.H.D and the Austrian Science Fund (FWF) [START Y275-B12] to A.L. Conflict of interest Orkide Ordu declares that she does not have any conflicts of interest. Alexandra Lusser declares that she does not have any conflicts of interest. Nynke H. Dekker declares that she does not have any conflicts of interest. Ethical approval This article does not contain any studies with human participants or animals performed by any of the authors.
http://www.ncbi.nlm.nih.gov/pubmed/23109894
1. Int J Mol Sci. 2012;13(9):11954-11973. doi: 10.3390/ijms130911954. Epub 2012 Sep 20. The emerging roles of ATP-dependent chromatin remodeling enzymes in nucleotide excision repair. Czaja W(1), Mao P(1), Smerdon MJ(1). Author information: (1)Department of Biochemistry and Biophysics, School of Molecular Biosciences, Washington State University, Pullman, WA, USA. DNA repair in eukaryotic cells takes place in the context of chromatin, where DNA, including damaged DNA, is tightly packed into nucleosomes and higher order chromatin structures. Chromatin intrinsically restricts accessibility of DNA repair proteins to the damaged DNA and impacts upon the overall rate of DNA repair. Chromatin is highly responsive to DNA damage and undergoes specific remodeling to facilitate DNA repair. How damaged DNA is accessed, repaired and restored to the original chromatin state, and how chromatin remodeling coordinates these processes in vivo, remains largely unknown. ATP-dependent chromatin remodelers (ACRs) are the master regulators of chromatin structure and dynamics. Conserved from yeast to humans, ACRs utilize the energy of ATP to reorganize packing of chromatin and control DNA accessibility by sliding, ejecting or restructuring nucleosomes. Several studies have demonstrated that ATP-dependent remodeling activity of ACRs plays important roles in coordination of spatio-temporal steps of different DNA repair pathways in chromatin. This review focuses on the role of ACRs in regulation of various aspects of nucleotide excision repair (NER) in the context of chromatin. We discuss current understanding of ATP-dependent chromatin remodeling by various subfamilies of remodelers and regulation of the NER pathway in vivo. DOI: 10.3390/ijms130911954 PMCID: PMC3472786 PMID: 23109894 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31794465
1. Continuum (Minneap Minn). 2019 Dec;25(6):1662-1681. doi: 10.1212/CON.0000000000000801. Facioscapulohumeral Muscular Dystrophies. Wagner KR. PURPOSE OF REVIEW: Facioscapulohumeral muscular dystrophy (FSHD) is a common muscular dystrophy affecting both pediatric and adult patients. This article reviews the phenotype and pathophysiology of the disease as well as the recent efforts in clinical outcome measures and clinical trials. RECENT FINDINGS: As the name implies, FSHD involves weakness of facial muscles, muscles that fix the scapula, and muscles overlying the humerus (biceps and triceps). The distinctive phenotype of FSHD occurs secondary to two different genetic mechanisms. FSHD type 1 (FSHD1) is due to a deletion on chromosome 4q, leading to hypomethylation and derepression of DUX4. FSHD type 2 (FSHD2) is due to mutations in SMCHD1 with resulting hypomethylation of the same subtelomeric region of chromosome 4q and derepression of DUX4. Understanding the central role of DUX4 has opened up the possibility of disease-modifying treatments. In preparation for clinical trials of novel agents, researchers are in the process of validating a number of clinical trial outcome measures including MRI, the 6-minute walk test, the FSHD Composite Outcome Measure, reachable workspace, electrical impedance myography, and the FSHD Health Index. SUMMARY: The treatment of FSHD is currently supportive only. While past clinical trials in FSHD have been largely disappointing, novel agents in development, including antisense oligonucleotides, gene therapy, and small molecules, hold promise for future meaningful therapies. DOI: 10.1212/CON.0000000000000801 PMID: 31794465 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34255854
1. Nucleic Acids Res. 2021 Aug 20;49(14):7925-7938. doi: 10.1093/nar/gkab553. RoboCOP: jointly computing chromatin occupancy profiles for numerous factors from chromatin accessibility data. Mitra S(1), Zhong J(2), Tran TQ(1), MacAlpine DM(2)(3)(4), Hartemink AJ(1)(2)(4). Author information: (1)Department of Computer Science, Duke University, Durham, NC 27708, USA. (2)Program in Computational Biology and Bioinformatics, Duke University, Durham, NC 27708, USA. (3)Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA. (4)Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA. Chromatin is a tightly packaged structure of DNA and protein within the nucleus of a cell. The arrangement of different protein complexes along the DNA modulates and is modulated by gene expression. Measuring the binding locations and occupancy levels of different transcription factors (TFs) and nucleosomes is therefore crucial to understanding gene regulation. Antibody-based methods for assaying chromatin occupancy are capable of identifying the binding sites of specific DNA binding factors, but only one factor at a time. In contrast, epigenomic accessibility data like MNase-seq, DNase-seq, and ATAC-seq provide insight into the chromatin landscape of all factors bound along the genome, but with little insight into the identities of those factors. Here, we present RoboCOP, a multivariate state space model that integrates chromatin accessibility data with nucleotide sequence to jointly compute genome-wide probabilistic scores of nucleosome and TF occupancy, for hundreds of different factors. We apply RoboCOP to MNase-seq and ATAC-seq data to elucidate the protein-binding landscape of nucleosomes and 150 TFs across the yeast genome, and show that our model makes better predictions than existing methods. We also compute a chromatin occupancy profile of the yeast genome under cadmium stress, revealing chromatin dynamics associated with transcriptional regulation. © The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research. DOI: 10.1093/nar/gkab553 PMCID: PMC8373080 PMID: 34255854 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35428982
1. Muscle Nerve. 2022 Jul;66(1):50-62. doi: 10.1002/mus.27558. Epub 2022 May 9. Randomized phase 2 study of ACE-083, a muscle-promoting agent, in facioscapulohumeral muscular dystrophy. Statland JM(1), Campbell C(2), Desai U(3), Karam C(4), Díaz-Manera J(5)(6)(7), Guptill JT(8), Korngut L(9), Genge A(10), Tawil RN(11), Elman L(12), Joyce NC(13), Wagner KR(14), Manousakis G(15), Amato AA(16), Butterfield RJ(17), Shieh PB(18), Wicklund M(19), Gamez J(20), Bodkin C(21), Pestronk A(22), Weihl CC(22), Vilchez-Padilla JJ(23)(6), Johnson NE(24), Mathews KD(25), Miller B(26), Leneus A(26), Fowler M(26), van de Rijn M(26), Attie KM(26). Author information: (1)Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, USA. (2)Department of Pediatrics and Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada. (3)Carolinas MDA Care Center, Atrium Health, Charlotte, North Carolina, USA. (4)Neuromuscular Division, Oregon Health & Science University, Portland, Oregon, USA. (5)Neuromuscular Diseases Unit, Neurology Department, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain. (6)Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain. (7)John Walton Muscular Dystrophy Research Centre, Newcastle University Translational and Clinical Research Institute, Newcastle, UK. (8)Department of Neurology, Duke University School of Medicine, Durham, North Carolina, USA. (9)University of Calgary, Calgary, Alberta, Canada. (10)Montreal Neurological Institute, Montreal, Quebec, Canada. (11)University of Rochester School of Medicine, Rochester, New York, USA. (12)University of Pennsylvania, Philadelphia, Pennsylvania, USA. (13)University of California Davis Medical Center, Davis, California, USA. (14)Johns Hopkins School of Medicine, Kennedy Krieger Institute, Baltimore, Maryland, USA. (15)Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA. (16)Brigham and Women's Hospital, Boston, Massachusetts, USA. (17)Departments of Neurology and Pediatrics, University of Utah, Salt Lake City, Utah, USA. (18)University of California Los Angeles, Los Angeles, California, USA. (19)University of Colorado, Aurora, Colorado, USA. (20)Department of Medicine, GMA Clinic, European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD) and Universitat Autònoma de Barcelona, Barcelona, Spain. (21)Indiana University School of Medicine, Indianapolis, Indiana, USA. (22)Washington University School of Medicine, St. Louis, Missouri, USA. (23)Hospital UIP La Fe, Neuromuscular Reference Centre, Valencia, Spain. (24)Virginia Commonwealth University, Richmond, Virginia, USA. (25)Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA. (26)Acceleron Pharma, Cambridge, Massachusetts, USA. INTRODUCTION/AIMS: Facioscapulohumeral muscular dystrophy (FSHD) is a slowly progressive muscular dystrophy without approved therapies. In this study we evaluated whether locally acting ACE-083 could safely increase muscle volume and improve functional outcomes in adults with FSHD. METHODS: Participants were at least 18 years old and had FSHD1/FSHD2. Part 1 was open label, ascending dose, assessing safety and tolerability (primary objective). Part 2 was randomized, double-blind for 6 months, evaluating ACE-083240 mg/muscle vs placebo injected bilaterally every 3 weeks in the biceps brachii (BB) or tibialis anterior (TA) muscles, followed by 6 months of open label. Magnetic resonance imaging measures included total muscle volume (TMV; primary objective), fat fraction (FF), and contractile muscle volume (CMV). Functional measures included 6-minute walk test, 10-meter walk/run, and 4-stair climb (TA group), and performance of upper limb midlevel/elbow score (BB group). Strength, patient-reported outcomes (PROs), and safety were also evaluated. RESULTS: Parts 1 and 2 enrolled 37 and 58 participants, respectively. Among 55 participants evaluable in Part 2, the least-squares mean (90% confidence interval, analysis of covariance) treatment difference for TMV was 16.4% (9.8%-23.0%) in the BB group (P < .0001) and 9.5% (3.2%-15.9%) in the TA group (P = .01). CMV increased significantly in the BB and TA groups and FF decreased in the TA group. There were no consistent improvements in functional or PRO measures in either group. The most common adverse events were mild or moderate injection-site reactions. DISCUSSION: Significant increases in TMV with ACE-083 vs placebo did not result in consistent functional or PRO improvements with up to 12 months of treatment. © 2022 The Authors. Muscle & Nerve published by Wiley Periodicals LLC. DOI: 10.1002/mus.27558 PMCID: PMC9321022 PMID: 35428982 [Indexed for MEDLINE] Conflict of interest statement: J.M. Statland received grant support from the NIH, MDA, FSHD Society, and the Friends of FSH Research; he is a consultant or has served on advisory boards for Dyne, Fulcrum, Acceleron, Avidity, Strongbridge, Sarepta, and Genzyme. U. Desai has served on advisory boards for Alexion, CSL Behring, Argenx, Akcea, and Stealth Biotherapeutics and has served on the speaker's bureau for Alexion. C. Karam has undertaken consulting or educational activities for Akcea, Alexion, Alnylam, Argenx, Biogen, CSL Behring, Medscape, and Sanofi Genzyme and has received research grants from Sanofi Genzyme and Akcea. J. Díaz‐Manera has served as a consultant or on advisory boards for Sanofi‐Genzyme, Amicus, Audentes, Sarepta, and Spark. He has also received industry grant support from Sanofi Genzyme and Boehringer Ingelheim. J.T. Guptill has served as a consultant or on advisory boards for Immunovant, Alexion, Momenta, Ra Pharma, Grifols, Argenx, Jacobus, Becton Dickinson, Cabaletta, Regeneron, and Piedmont Pharmaceuticals and receives industry grant support from UCB for a fellowship training grant. A. Genge serves as a consultant for Mitsubishi Tanabe Pharma America, Sanofi Genzyme, AL‐S Pharma, AB Sciences, Biogen, Novartis, CSL Behring, Anavex, AveXis, Alexion, Wave Life Sciences, Revalesio, Roche, Cytokinetics, Orion, Akcea, Clene, Bayshore, and QurAlis. She participates as CRU Medical Director, PI, or sub‐PI on trials sponsored by AB Sciences, AL‐S Pharma, Acceleron, Amicus, Alnylam, Bioblast, Biogen, BMS, Boston Biomedical Cytokinetics, Sanofi Genzyme, Grifols, Ionis, Eli Lilly, Mallinckrodt, MedImmune, Novartis, Orion, Orphazyme, Pfizer, Ra Pharmaceuticals, Roche, Teva, and UCB. R.N. Tawil serves as an advisory board member or consultant for Acceleron Pharma, Fulcrum Therapeutics, MT Pharma, and Arrowhead Pharma. L. Elman has served on advisory boards for Roche/Genentech and Biogen and received royalties from UpToDate (Wolters Kluwer). K.R. Wagner has served on advisory boards or consulted for AskBio, Dyne, Arrowhead Pharma, Catabasis, Santhera, and Vita. G. Manousakis has served on advisory boards for Stealth Biotherapeutics and Argenx. A.A. Amato is an associate editor for Neurology and has served as a medical consultant or on advisory boards for Sarepta, Alexion, and Serono; he received royalties from UpToDate (Wolters Kluwer) and Harrison's Principles of Internal Medicine, Neuromuscular Disorders, 2nd ed. R.J. Butterfield is receiving funding via contracts for clinical trials from AveXis, PTC Therapeutics, Sarepta Therapeutics, Pfizer, Biogen, Capricorn, and Catabasis; he serves on scientific advisory boards for Sarepta Therapeutics, Biogen, AveXis, and Pfizer. M. Wicklund has received research funding from the NIH, MDA, Acceleron, Alexion, Baxalta, ML Bio, Orphazyme, and Sarepta Therapeutics and has served on advisory boards or in consultation for Affinia, Amicus, ML Bio, Sanofi, and Sarepta. J. Gamez has received grant funding from Fondo de Investigación Sanitaria (FIS‐FEDER) (grants PI16/01673 and PI19/00593). N.E. Johnson has received grant funding from the NINDS (4K23NS091511; R01NS104010), CDC (DD19‐002), and the FDA (7R01FD006071‐02); he receives royalties from the Congenital and Childhood Onset Myotonic Dystrophy Health Index and the Charcot‐Marie‐Tooth Health Index; receives research funds from Dyne, AveXis, CSL Behring, Vertex Pharmaceuticals, Fulcrum Therapeutics, ML Bio, Sarepta, and Acceleron Pharma; and has provided consultation for AveXis, AMO Pharma, Strongbridge BioPharma, Acceleron Pharma, Fulcrum Therapeutics, Dyne, Avidity, and Vertex Pharmaceuticals. B. Miller, A. Leneus, M. Fowler, M. van de Rijn, and K. Attie were employed by Acceleron Pharma during the study and had stock ownership and/or options. The remaining authors declare no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/34016622
1. Cancer Res. 2021 Aug 15;81(16):4174-4182. doi: 10.1158/0008-5472.CAN-20-4010. Epub 2021 May 20. Implications of Enhancer Transcription and eRNAs in Cancer. Adhikary S(1)(2), Roy S(2), Chacon J(3), Gadad SS(4)(5)(6)(7), Das C(8)(9). Author information: (1)Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India. (2)Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India. (3)Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas. (4)Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas. [email protected] [email protected]. (5)Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas. (6)Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas. (7)Cecil H. and Ida Green Center for Reproductive Biology Sciences, Department of Obstetrics and Gynaecology, University of Texas Southwestern Medical Center, Dallas, Texas. (8)Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India. [email protected] [email protected]. (9)Homi Bhaba National Institute, Mumbai, Maharashtra, India. Despite extensive progress in developing anticancer therapies, therapy resistance remains a major challenge that promotes disease relapse. The changes that lead to therapy resistance can be intrinsically present or may be initiated during treatment. Genetic and epigenetic heterogeneity in tumors make it more challenging to deal with therapy resistance. Recent advances in genome-wide analyses have revealed that the deregulation of distal gene regulatory elements, such as enhancers, appears in several pathophysiological conditions, including cancer. Beyond the conventional function of enhancers in recruiting transcription factors to gene promoters, enhancer elements are also transcribed into noncoding RNAs known as enhancer RNAs (eRNA). Accumulating evidence suggests that uncontrolled enhancer activity with aberrant eRNA expression promotes oncogenesis. Interestingly, tissue-specific, transcribed eRNAs from active enhancers can serve as potential therapeutic targets or biomarkers in several cancer types. This review provides a comprehensive overview of the mechanisms of enhancer transcription and eRNAs as well as their potential roles in cancer and drug resistance. ©2021 American Association for Cancer Research. DOI: 10.1158/0008-5472.CAN-20-4010 PMID: 34016622 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35277481
1. Cell Death Dis. 2022 Mar 11;13(3):228. doi: 10.1038/s41419-022-04673-4. Super-enhancers and novel therapeutic targets in colorectal cancer. Liu Q(1)(2), Guo L(1)(3), Lou Z(1)(3), Xiang X(4)(5), Shao J(6)(7)(8). Author information: (1)Department of Pathology & Pathophysiology, and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. (2)Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China. (3)Key Laboratory of Disease Proteomics of Zhejiang Province, Key Laboratory of Cancer Prevention and Intervention of China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China. (4)Department of Pathology & Pathophysiology, and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. [email protected]. (5)Key Laboratory of Disease Proteomics of Zhejiang Province, Key Laboratory of Cancer Prevention and Intervention of China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China. [email protected]. (6)Department of Pathology & Pathophysiology, and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. [email protected]. (7)Key Laboratory of Disease Proteomics of Zhejiang Province, Key Laboratory of Cancer Prevention and Intervention of China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China. [email protected]. (8)Cancer Center, Zhejiang University, Hangzhou, China. [email protected]. Transcription factors, cofactors, chromatin regulators, and transcription apparatuses interact with transcriptional regulatory elements, including promoters, enhancers, and super-enhancers (SEs), to coordinately regulate the transcription of target genes and thereby control cell behaviors. Among these transcriptional regulatory components and related elements, SEs often play a central role in determining cell identity and tumor initiation and progression. Therefore, oncogenic SEs, which are generated within cancer cells in oncogenes and other genes important in tumor pathogenesis, have emerged as attractive targets for novel cancer therapeutic strategies in recent years. Herein, we review the identification, formation and activation modes, and regulatory mechanisms for downstream genes and pathways of oncogenic SEs. We also review the therapeutic strategies and compounds targeting oncogenic SEs in colorectal cancer and other malignancies. © 2022. The Author(s). DOI: 10.1038/s41419-022-04673-4 PMCID: PMC8917125 PMID: 35277481 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/33934105
1. Oncogene. 2021 May;40(20):3475-3491. doi: 10.1038/s41388-021-01793-7. Epub 2021 May 1. Enhancer rewiring in tumors: an opportunity for therapeutic intervention. Richart L(1)(2), Bidard FC(3)(4)(5), Margueron R(6)(7). Author information: (1)Institut Curie, Paris Sciences et Lettres Research University, Paris, France. (2)INSERM U934/CNRS UMR3215, Paris, France. (3)Department of Medical Oncology, Institut Curie, Saint-Cloud, Paris, France. (4)Circulating Tumor Biomarkers Laboratory, SIRIC2 Institut Curie, Paris, France. (5)UVSQ, Université Paris-Saclay, Saint-Cloud, Paris, France. (6)Institut Curie, Paris Sciences et Lettres Research University, Paris, France. [email protected]. (7)INSERM U934/CNRS UMR3215, Paris, France. [email protected]. Enhancers are cis-regulatory sequences that fine-tune expression of their target genes in a spatiotemporal manner. They are recognized by sequence-specific transcription factors, which in turn recruit transcriptional coactivators that facilitate transcription by promoting assembly and activation of the basal transcriptional machinery. Their functional importance is underscored by the fact that they are often the target of genetic and nongenetic events in human disease that disrupt their sequence, interactome, activation potential, and/or chromatin environment. Dysregulation of transcription and addiction to transcriptional effectors that interact with and modulate enhancer activity are common features of cancer cells and are amenable to therapeutic intervention. Here, we discuss the current knowledge on enhancer biology, the broad spectrum of mechanisms that lead to their malfunction in tumor cells, and recent progress in developing drugs that efficaciously target their dependencies. DOI: 10.1038/s41388-021-01793-7 PMID: 33934105 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33299103
1. NPJ Precis Oncol. 2020 Nov 19;4(1):31. doi: 10.1038/s41698-020-00137-0. Super-enhancer in prostate cancer: transcriptional disorders and therapeutic targets. Chen X(1), Ma Q(1), Shang Z(2), Niu Y(3). Author information: (1)Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China. (2)Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China. [email protected]. (3)Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China. [email protected]. Abnormal activity of oncogenic and tumor-suppressor signaling pathways contributes to cancer and cancer risk in humans. Transcriptional dysregulation of these pathways is commonly associated with tumorigenesis and the development of cancer. Genetic and epigenetic alterations may mediate dysregulated transcriptional activity. One of the most important epigenetic alternations is the non-coding regulatory element, which includes both enhancers and super-enhancers (SEs). SEs, characterized as large clusters of enhancers with aberrant high levels of transcription factor binding, have been considered as key drivers of gene expression in controlling and maintaining cancer cell identity. In cancer cells, oncogenes acquire SEs and the cancer phenotype relies on these abnormal transcription programs driven by SEs, which leads to cancer cells often becoming addicted to the SEs-related transcription programs, including prostate cancer. Here, we summarize recent findings of SEs and SEs-related gene regulation in prostate cancer and review the potential pharmacological inhibitors in basic research and clinical trials. DOI: 10.1038/s41698-020-00137-0 PMCID: PMC7677538 PMID: 33299103 Conflict of interest statement: The authors declare no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/25473436
1. Genome Med. 2014 Sep 23;6(9):77. doi: 10.1186/s13073-014-0077-3. eCollection 2014. Enhancer alterations in cancer: a source for a cell identity crisis. Kron KJ(1), Bailey SD(1), Lupien M(2). Author information: (1)The Princess Margaret Cancer Centre - University Health Network, Toronto, ON M5G 1 L7 Canada ; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1 L7 Canada. (2)The Princess Margaret Cancer Centre - University Health Network, Toronto, ON M5G 1 L7 Canada ; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1 L7 Canada ; Ontario Institute for Cancer Research, Toronto, ON M5G 0A3 Canada. Enhancers are selectively utilized to orchestrate gene expression programs that first govern pluripotency and then proceed to highly specialized programs required for the process of cellular differentiation. Whereas gene-proximal promoters are typically active across numerous cell types, distal enhancer activation is cell-type-specific and central to cell fate determination, thereby accounting for cell identity. Recent studies have highlighted the diversity of enhancer usage, cataloguing millions of such elements in the human genome. The disruption of enhancer activity, through genetic or epigenetic alterations, can impact cell-type-specific functions, resulting in a wide range of pathologies. In cancer, these alterations can promote a 'cell identity crisis', in which enhancers associated with oncogenes and multipotentiality are activated, while those promoting cell fate commitment are inactivated. Overall, these alterations favor an undifferentiated cellular phenotype. Here, we review the current knowledge regarding the role of enhancers in normal cell function, and discuss how genetic and epigenetic changes in enhancer elements potentiate oncogenesis. In addition, we discuss how understanding the mechanisms regulating enhancer activity can inform therapeutic opportunities in cancer cells and highlight key challenges that remain in understanding enhancer biology as it relates to oncology. DOI: 10.1186/s13073-014-0077-3 PMCID: PMC4254433 PMID: 25473436
http://www.ncbi.nlm.nih.gov/pubmed/35750313
1. Biochim Biophys Acta Gene Regul Mech. 2022 Aug;1865(6):194839. doi: 10.1016/j.bbagrm.2022.194839. Epub 2022 Jun 22. Multifaceted regulation of enhancers in cancer. Xiao Q(1), Xiao Y(1), Li LY(1), Chen MK(2), Wu M(3). Author information: (1)Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430072, China. (2)Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430072, China. Electronic address: [email protected]. (3)Frontier Science Center for Immunology and Metabolism, Hubei Key Laboratory of Cell Homeostasis, Hubei Key Laboratory of Developmentally Originated Disease, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430072, China. Electronic address: [email protected]. Enhancer is one kind of cis-elements regulating gene transcription, whose activity is tightly controlled by epigenetic enzymes and histone modifications. Active enhancers are classified into typical enhancers, super-enhancers and over-active enhancers, according to the enrichment and location of histone modifications. Epigenetic factors control the level of histone modifications on enhancers to determine their activity, such as histone methyltransferases and acetylases. Transcription factors, cofactors and mediators co-operate together and are required for enhancer functions. In turn, abnormalities in these trans-acting factors affect enhancer activity. Recent studies have revealed enhancer dysregulation as one of the important features for cancer. Variations in enhancer regions and mutations of enhancer regulatory genes are frequently observed in cancer cells, and altering the activity of onco-enhancers is able to repress oncogene expression, and suppress tumorigenesis and metastasis. Here we summarize the recent discoveries about enhancer regulation in cancer and discuss their potential application in diagnosis and treatment. Copyright © 2022. Published by Elsevier B.V. DOI: 10.1016/j.bbagrm.2022.194839 PMID: 35750313 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27364481
1. Nat Rev Cancer. 2016 Aug;16(8):483-93. doi: 10.1038/nrc.2016.62. Epub 2016 Jul 1. The role of enhancers in cancer. Sur I(1), Taipale J(1)(2). Author information: (1)Division of Functional Genomics and Systems Biology, Department of Medical Biochemistry and Biophysics, and Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm SE-171 77, Sweden. (2)Genome-Scale Biology Program, University of Helsinki, Biomedicum, PO Box 63, Helsinki 00014, Finland. Enhancer elements function as the logic gates of the genetic regulatory circuitry. One of their most important functions is the integration of extracellular signals with intracellular cell fate information to generate cell type-specific transcriptional responses. Mutations occurring in cancer often misregulate enhancers that normally control the signal-dependent expression of growth-related genes. This misregulation can result from trans-acting mechanisms, such as activation of the transcription factors or epigenetic regulators that control enhancer activity, or can be caused in cis by direct mutations that alter the activity of the enhancer or its target gene specificity. These processes can generate tumour type-specific super-enhancers and establish a 'locked' gene regulatory state that drives the uncontrolled proliferation of cancer cells. Here, we review the role of enhancers in cancer, and their potential as therapeutic targets. DOI: 10.1038/nrc.2016.62 PMID: 27364481 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35911061
1. Epigenet Insights. 2022 Jul 26;15:25168657221115656. doi: 10.1177/25168657221115656. eCollection 2022. Emerging Themes in Mechanisms of Tumorigenesis by SWI/SNF Subunit Mutation. Jones CA(1), Tansey WP(2)(3), Weissmiller AM(1). Author information: (1)Department of Biology, Middle Tennessee State University, Murfreesboro, TN, USA. (2)Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA. (3)Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA. The SWI/SNF chromatin remodeling complex uses the energy of ATP hydrolysis to alter contacts between DNA and nucleosomes, allowing regions of the genome to become accessible for biological processes such as transcription. The SWI/SNF chromatin remodeler is also one of the most frequently altered protein complexes in cancer, with upwards of 20% of all cancers carrying mutations in a SWI/SNF subunit. Intense studies over the last decade have probed the molecular events associated with SWI/SNF dysfunction in cancer and common themes are beginning to emerge in how tumor-associated SWI/SNF mutations promote malignancy. In this review, we summarize current understanding of SWI/SNF complexes, their alterations in cancer, and what is known about the impact of these mutations on tumor-relevant transcriptional events. We discuss how enhancer dysregulation is a common theme in SWI/SNF mutant cancers and describe how resultant alterations in enhancer and super-enhancer activity conspire to block development and differentiation while promoting stemness and self-renewal. We also identify a second emerging theme in which SWI/SNF perturbations intersect with potent oncoprotein transcription factors AP-1 and MYC to drive malignant transcriptional programs. © The Author(s) 2022. DOI: 10.1177/25168657221115656 PMCID: PMC9329810 PMID: 35911061 Conflict of interest statement: Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
http://www.ncbi.nlm.nih.gov/pubmed/35170113
1. Cell Prolif. 2022 Apr;55(4):e13202. doi: 10.1111/cpr.13202. Epub 2022 Feb 16. Enhancer RNA: What we know and what we can achieve. Han Z(1), Li W(1). Author information: (1)Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China. Enhancers are important cis-acting elements that can regulate gene transcription and cell fate alongside promoters. In fact, many human cancers and diseases are associated with the malfunction of enhancers. Recent studies have shown that enhancers can produce enhancer RNAs (eRNAs) by RNA polymerase II. In this review, we discuss eRNA production, characteristics, functions and mechanics. eRNAs can determine chromatin accessibility, histone modification and gene expression by constructing a 'chromatin loop', thereby bringing enhancers to their target gene. eRNA can also be involved in the phase separation with enhancers and other proteins. eRNAs are abundant, and importantly, tissue-specific in tumours, various diseases and stem cells; thus, eRNAs can be a potential target for disease diagnosis and treatment. As eRNA is produced from the active transcription of enhancers and is involved in the regulation of cell fate, its manipulation will influence cell function, and therefore, it can be a new target for biological therapy. © 2022 The Authors. Cell Proliferation Published by John Wiley & Sons Ltd. DOI: 10.1111/cpr.13202 PMCID: PMC9055912 PMID: 35170113 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/31916476
1. RNA Biol. 2020 Nov;17(11):1550-1559. doi: 10.1080/15476286.2020.1712895. Epub 2020 Jan 19. Enhancer RNAs in cancer: regulation, mechanisms and therapeutic potential. Lee JH(1), Xiong F(1), Li W(1)(2). Author information: (1)Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center , Houston, TX, USA. (2)Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth , Houston, TX, USA. Enhancers are distal genomic elements critical for gene regulation and cell identify control during development and diseases. Many human cancers were found to associate with enhancer malfunction, due to genetic and epigenetic alterations, which in some cases directly drive tumour growth. Conventionally, enhancers are known to provide DNA binding motifs to recruit transcription factors (TFs) and to control target genes. However, recent progress found that most, if not all, active enhancers pervasively transcribe noncoding RNAs that are referred to as enhancer RNAs (eRNAs). Increasing evidence points to functional roles of at least a subset of eRNAs in gene regulation in both normal and cancer cells, adding new insights into the action mechanisms of enhancers. eRNA expression was observed to be widespread but also specific to tumour types and individual patients, serving as opportunities to exploit them as potential diagnosis markers or therapeutic targets. In this review, we discuss the brief history of eRNA research, their functional mechanisms and importance in cancer gene regulation, as well as their therapeutic and diagnostic values in cancer. We propose that further studies of eRNAs in cancer will offer a promising 'eRNA targeted therapy' for human cancer intervention. DOI: 10.1080/15476286.2020.1712895 PMCID: PMC7567500 PMID: 31916476 [Indexed for MEDLINE] Conflict of interest statement: No potential conflict of interest was reported by the authors.
http://www.ncbi.nlm.nih.gov/pubmed/35967935
1. Nagoya J Med Sci. 2022 May;84(2):216-229. doi: 10.18999/nagjms.84.2.216. The molecular understanding of super-enhancer dysregulation in cancer. Yoshino S(1), Suzuki HI(1)(2). Author information: (1)Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan. (2)Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan. Abnormalities in the regulation of gene expression are associated with various pathological conditions. Among the distal regulatory elements in the genome, the activation of target genes by enhancers plays a central role in the formation of cell type-specific gene expression patterns. Super-enhancers are a subclass of enhancers that frequently contain multiple enhancer-like elements and are characterized by dense binding of master transcription factors and Mediator complexes and high signals of active histone marks. Super-enhancers have been studied in detail as important regulatory regions that control cell identity and contribute to the pathogenesis of diverse diseases. In cancer, super-enhancers have multifaceted roles by activating various oncogenes and other cancer-related genes and shaping characteristic gene expression patterns in cancer cells. Alterations in super-enhancer activities in cancer involve multiple mechanisms, including the dysregulation of transcription factors and the super-enhancer-associated genomic abnormalities. The study of super-enhancers could contribute to the identification of effective biomarkers and the development of cancer therapeutics targeting transcriptional addiction. In this review, we summarize the roles of super-enhancers in cancer biology, with a particular focus on hematopoietic malignancies, in which multiple super-enhancer alteration mechanisms have been reported. DOI: 10.18999/nagjms.84.2.216 PMCID: PMC9350580 PMID: 35967935 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35406623
1. Cancers (Basel). 2022 Apr 6;14(7):1852. doi: 10.3390/cancers14071852. Enhancer RNA Transcription Is Essential for a Novel CSF1 Enhancer in Triple-Negative Breast Cancer. Lewis MW(1), Wisniewska K(1), King CM(1), Li S(1), Coffey A(1), Kelly MR(1)(2), Regner MJ(1)(2), Franco HL(1)(2)(3). Author information: (1)The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. (2)Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. (3)The Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. Enhancers are critical regulatory elements in the genome that help orchestrate spatiotemporal patterns of gene expression during development and normal physiology. In cancer, enhancers are often rewired by various genetic and epigenetic mechanisms for the activation of oncogenes that lead to initiation and progression. A key feature of active enhancers is the production of non-coding RNA molecules called enhancer RNAs, whose functions remain unknown but can be used to specify active enhancers de novo. Using a combination of eRNA transcription and chromatin modifications, we have identified a novel enhancer located 30 kb upstream of Colony Stimulating Factor 1 (CSF1). Notably, CSF1 is implicated in the progression of breast cancer, is overexpressed in triple-negative breast cancer (TNBC) cell lines, and its enhancer is primarily active in TNBC patient tumors. Genomic deletion of the enhancer (via CRISPR/Cas9) enabled us to validate this regulatory element as a bona fide enhancer of CSF1 and subsequent cell-based assays revealed profound effects on cancer cell proliferation, colony formation, and migration. Epigenetic silencing of the enhancer via CRISPR-interference assays (dCas9-KRAB) coupled to RNA-sequencing, enabled unbiased identification of additional target genes, such as RSAD2, that are predictive of clinical outcome. Additionally, we repurposed the RNA-guided RNA-targeting CRISPR-Cas13 machinery to specifically degrade the eRNAs transcripts produced at this enhancer to determine the consequences on CSF1 mRNA expression, suggesting a post-transcriptional role for these non-coding transcripts. Finally, we test our eRNA-dependent model of CSF1 enhancer function and demonstrate that our results are extensible to other forms of cancer. Collectively, this work describes a novel enhancer that is active in the TNBC subtype, which is associated with cellular growth, and requires eRNA transcripts for proper enhancer function. These results demonstrate the significant impact of enhancers in cancer biology and highlight their potential as tractable targets for therapeutic intervention. DOI: 10.3390/cancers14071852 PMCID: PMC8997997 PMID: 35406623 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/30885876
1. Pharmacol Ther. 2019 Jul;199:129-138. doi: 10.1016/j.pharmthera.2019.02.014. Epub 2019 Mar 16. Super-enhancers in cancer. Thandapani P(1). Author information: (1)Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA. Electronic address: [email protected]. Cancer is fueled by the aberrant activity of oncogenic and tumor suppressive pathways. Transcriptional dysregulation of these pathways play a major role both in the genesis and development of cancer. Dysregulation of transcriptional programs can be mediated by genetic and epigenetic alterations targeting both protein coding genes and non-coding regulatory elements like enhancers and super-enhancers. Super-enhancers, characterized as large clusters of enhancers in close proximity, have been identified as essential oncogenic drivers required for the maintenance of cancer cell identity. As a result, cancer cells are often addicted to the super-enhancer driven transcriptional programs. Furthermore, pharmacological inhibitors targeting key components of super-enhancer assembly and activation have shown great promise in reducing tumor growth and proliferation in several pre-clinical tumor models. This article reviews the current understanding of super-enhancer assembly and activation, the different mechanisms by which cancer cells acquire oncogenic super-enhancers and, finally, the potential of targeting super-enhancers as future therapeutics. Copyright © 2019 Elsevier Inc. All rights reserved. DOI: 10.1016/j.pharmthera.2019.02.014 PMID: 30885876 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36387198
1. Front Oncol. 2022 Oct 27;12:1036648. doi: 10.3389/fonc.2022.1036648. eCollection 2022. Super-enhancers in esophageal carcinoma: Transcriptional addictions and therapeutic strategies. Shi Y(1)(2), Wang M(3), Liu D(1)(2), Ullah S(1)(2), Ma X(4), Yang H(1)(2), Liu B(1)(2). Author information: (1)Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. (2)State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China. (3)Academy of Medical Sciences Zhengzhou University, Zhengzhou, China. (4)Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China. The tumorigenesis of esophageal carcinoma arises from transcriptional dysregulation would become exceptionally dependent on specific regulators of gene expression, which could be preferentially attributed to the larger non-coding cis-regulatory elements, i.e. super-enhancers (SEs). SEs, large genomic regulatory entity in close genomic proximity, are underpinned by control cancer cell identity. As a consequence, the transcriptional addictions driven by SEs could offer an Achilles' heel for molecular treatments on patients of esophageal carcinoma and other types of cancer as well. In this review, we summarize the recent findings about the oncogenic SEs upon which esophageal cancer cells depend, and discuss why SEs could be seen as the hallmark of cancer, how transcriptional dependencies driven by SEs, and what opportunities could be supplied based on this cancer-specific SEs. Copyright © 2022 Shi, Wang, Liu, Ullah, Ma, Yang and Liu. DOI: 10.3389/fonc.2022.1036648 PMCID: PMC9647064 PMID: 36387198 Conflict of interest statement: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/34210001
1. Cancers (Basel). 2021 Jun 29;13(13):3270. doi: 10.3390/cancers13133270. Enhancing B-Cell Malignancies-On Repurposing Enhancer Activity towards Cancer. Kasprzyk ME(1), Sura W(1), Dzikiewicz-Krawczyk A(1). Author information: (1)Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznań, Poland. B-cell lymphomas and leukemias derive from B cells at various stages of maturation and are the 6th most common cancer-related cause of death. While the role of several oncogenes and tumor suppressors in the pathogenesis of B-cell neoplasms was established, recent research indicated the involvement of non-coding, regulatory sequences. Enhancers are DNA elements controlling gene expression in a cell type- and developmental stage-specific manner. They ensure proper differentiation and maturation of B cells, resulting in production of high affinity antibodies. However, the activity of enhancers can be redirected, setting B cells on the path towards cancer. In this review we discuss different mechanisms through which enhancers are exploited in malignant B cells, from the well-studied translocations juxtaposing oncogenes to immunoglobulin loci, through enhancer dysregulation by sequence variants and mutations, to enhancer hijacking by viruses. We also highlight the potential of therapeutic targeting of enhancers as a direction for future investigation. DOI: 10.3390/cancers13133270 PMCID: PMC8269369 PMID: 34210001 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35454885
1. Cancers (Basel). 2022 Apr 14;14(8):1978. doi: 10.3390/cancers14081978. Enhancer RNAs (eRNAs) in Cancer: The Jacks of All Trades. Napoli S(1), Munz N(1), Guidetti F(1), Bertoni F(1)(2). Author information: (1)Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland. (2)Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland. Enhancer RNAs (eRNAs) are non-coding RNAs (ncRNAs) transcribed in enhancer regions. They play an important role in transcriptional regulation, mainly during cellular differentiation. eRNAs are tightly tissue- and cell-type specific and are induced by specific stimuli, activating promoters of target genes in turn. eRNAs usually have a very short half-life but in some cases, once activated, they can be stably expressed and acquire additional functions. Due to their critical role, eRNAs are often dysregulated in cancer and growing number of interactions with chromatin modifiers, transcription factors, and splicing machinery have been described. Enhancer activation and eRNA transcription have particular relevance also in inflammatory response, placing the eRNAs at the interplay between cancer and immune cells. Here, we summarize all the possible molecular mechanisms recently reported in association with eRNAs activity. DOI: 10.3390/cancers14081978 PMCID: PMC9030334 PMID: 35454885 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/29625054
1. Cell. 2018 Apr 5;173(2):386-399.e12. doi: 10.1016/j.cell.2018.03.027. A Pan-Cancer Analysis of Enhancer Expression in Nearly 9000 Patient Samples. Chen H(1), Li C(2), Peng X(1), Zhou Z(3), Weinstein JN(3); Cancer Genome Atlas Research Network; Liang H(4). Collaborators: Caesar-Johnson SJ, Demchok JA, Felau I, Kasapi M, Ferguson ML, Hutter CM, Sofia HJ, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Zhang JJ, Chudamani S, Liu J, Lolla L, Naresh R, Pihl T, Sun Q, Wan Y, Wu Y, Cho J, DeFreitas T, Frazer S, Gehlenborg N, Getz G, Heiman DI, Kim J, Lawrence MS, Lin P, Meier S, Noble MS, Saksena G, Voet D, Zhang H, Bernard B, Chambwe N, Dhankani V, Knijnenburg T, Kramer R, Leinonen K, Liu Y, Miller M, Reynolds S, Shmulevich I, Thorsson V, Zhang W, Akbani R, Broom BM, Hegde AM, Ju Z, Kanchi RS, Korkut A, Li J, Liang H, Ling S, Liu W, Lu Y, Mills GB, Ng KS, Rao A, Ryan M, Wang J, Weinstein JN, Zhang J, Abeshouse A, Armenia J, Chakravarty D, Chatila WK, de Bruijn I, Gao J, Gross BE, Heins ZJ, Kundra R, La K, Ladanyi M, Luna A, Nissan MG, Ochoa A, Phillips SM, Reznik E, Sanchez-Vega F, Sander C, Schultz N, Sheridan R, Sumer SO, Sun Y, Taylor BS, Wang J, Zhang H, Anur P, Peto M, Spellman P, Benz C, Stuart JM, Wong CK, Yau C, Hayes DN, Parker JS, Wilkerson MD, Ally A, Balasundaram M, Bowlby R, Brooks D, Carlsen R, Chuah E, Dhalla N, Holt R, Jones SJM, Kasaian K, Lee D, Ma Y, Marra MA, Mayo M, Moore RA, Mungall AJ, Mungall K, Robertson AG, Sadeghi S, Schein JE, Sipahimalani P, Tam A, Thiessen N, Tse K, Wong T, Berger AC, Beroukhim R, Cherniack AD, Cibulskis C, Gabriel SB, Gao GF, Ha G, Meyerson M, Schumacher SE, Shih J, Kucherlapati MH, Kucherlapati RS, Baylin S, Cope L, Danilova L, Bootwalla MS, Lai PH, Maglinte DT, Van Den Berg DJ, Weisenberger DJ, Auman JT, Balu S, Bodenheimer T, Fan C, Hoadley KA, Hoyle AP, Jefferys SR, Jones CD, Meng S, Mieczkowski PA, Mose LE, Perou AH, Perou CM, Roach J, Shi Y, Simons JV, Skelly T, Soloway MG, Tan D, Veluvolu U, Fan H, Hinoue T, Laird PW, Shen H, Zhou W, Bellair M, Chang K, Covington K, Creighton CJ, Dinh H, Doddapaneni H, Donehower LA, Drummond J, Gibbs RA, Glenn R, Hale W, Han Y, Hu J, Korchina V, Lee S, Lewis L, Li W, Liu X, Morgan M, Morton D, Muzny D, Santibanez J, Sheth M, Shinbrot E, Wang L, Wang M, Wheeler DA, Xi L, Zhao F, Hess J, Appelbaum EL, Bailey M, Cordes MG, Ding L, Fronick CC, Fulton LA, Fulton RS, Kandoth C, Mardis ER, McLellan MD, Miller CA, Schmidt HK, Wilson RK, Crain D, Curley E, Gardner J, Lau K, Mallery D, Morris S, Paulauskis J, Penny R, Shelton C, Shelton T, Sherman M, Thompson E, Yena P, Bowen J, Gastier-Foster JM, Gerken M, Leraas KM, Lichtenberg TM, Ramirez NC, Wise L, Zmuda E, Corcoran N, Costello T, Hovens C, Carvalho AL, de Carvalho AC, Fregnani JH, Longatto-Filho A, Reis RM, Scapulatempo-Neto C, Silveira HCS, Vidal DO, Burnette A, Eschbacher J, Hermes B, Noss A, Singh R, Anderson ML, Castro PD, Ittmann M, Huntsman D, Kohl B, Le X, Thorp R, Andry C, Duffy ER, Lyadov V, Paklina O, Setdikova G, Shabunin A, Tavobilov M, McPherson C, Warnick R, Berkowitz R, Cramer D, Feltmate C, Horowitz N, Kibel A, Muto M, Raut CP, Malykh A, Barnholtz-Sloan JS, Barrett W, Devine K, Fulop J, Ostrom QT, Shimmel K, Wolinsky Y, Sloan AE, De Rose A, Giuliante F, Goodman M, Karlan BY, Hagedorn CH, Eckman J, Harr J, Myers J, Tucker K, Zach LA, Deyarmin B, Hu H, Kvecher L, Larson C, Mural RJ, Somiari S, Vicha A, Zelinka T, Bennett J, Iacocca M, Rabeno B, Swanson P, Latour M, Lacombe L, Têtu B, Bergeron A, McGraw M, Staugaitis SM, Chabot J, Hibshoosh H, Sepulveda A, Su T, Wang T, Potapova O, Voronina O, Desjardins L, Mariani O, Roman-Roman S, Sastre X, Stern MH, Cheng F, Signoretti S, Berchuck A, Bigner D, Lipp E, Marks J, McCall S, McLendon R, Secord A, Sharp A, Behera M, Brat DJ, Chen A, Delman K, Force S, Khuri F, Magliocca K, Maithel S, Olson JJ, Owonikoko T, Pickens A, Ramalingam S, Shin DM, Sica G, Van Meir EG, Zhang H, Eijckenboom W, Gillis A, Korpershoek E, Looijenga L, Oosterhuis W, Stoop H, van Kessel KE, Zwarthoff EC, Calatozzolo C, Cuppini L, Cuzzubbo S, DiMeco F, Finocchiaro G, Mattei L, Perin A, Pollo B, Chen C, Houck J, Lohavanichbutr P, Hartmann A, Stoehr C, Stoehr R, Taubert H, Wach S, Wullich B, Kycler W, Murawa D, Wiznerowicz M, Chung K, Edenfield WJ, Martin J, Baudin E, Bubley G, Bueno R, De Rienzo A, Richards WG, Kalkanis S, Mikkelsen T, Noushmehr H, Scarpace L, Girard N, Aymerich M, Campo E, Giné E, Guillermo AL, Van Bang N, Hanh PT, Phu BD, Tang Y, Colman H, Evason K, Dottino PR, Martignetti JA, Gabra H, Juhl H, Akeredolu T, Stepa S, Hoon D, Ahn K, Kang KJ, Beuschlein F, Breggia A, Birrer M, Bell D, Borad M, Bryce AH, Castle E, Chandan V, Cheville J, Copland JA, Farnell M, Flotte T, Giama N, Ho T, Kendrick M, Kocher JP, Kopp K, Moser C, Nagorney D, O'Brien D, O'Neill BP, Patel T, Petersen G, Que F, Rivera M, Roberts L, Smallridge R, Smyrk T, Stanton M, Thompson RH, Torbenson M, Yang JD, Zhang L, Brimo F, Ajani JA, Gonzalez AMA, Behrens C, Bondaruk J, Broaddus R, Czerniak B, Esmaeli B, Fujimoto J, Gershenwald J, Guo C, Lazar AJ, Logothetis C, Meric-Bernstam F, Moran C, Ramondetta L, Rice D, Sood A, Tamboli P, Thompson T, Troncoso P, Tsao A, Wistuba I, Carter C, Haydu L, Hersey P, Jakrot V, Kakavand H, Kefford R, Lee K, Long G, Mann G, Quinn M, Saw R, Scolyer R, Shannon K, Spillane A, Stretch J, Synott M, Thompson J, Wilmott J, Al-Ahmadie H, Chan TA, Ghossein R, Gopalan A, Levine DA, Reuter V, Singer S, Singh B, Tien NV, Broudy T, Mirsaidi C, Nair P, Drwiega P, Miller J, Smith J, Zaren H, Park JW, Hung NP, Kebebew E, Linehan WM, Metwalli AR, Pacak K, Pinto PA, Schiffman M, Schmidt LS, Vocke CD, Wentzensen N, Worrell R, Yang H, Moncrieff M, Goparaju C, Melamed J, Pass H, Botnariuc N, Caraman I, Cernat M, Chemencedji I, Clipca A, Doruc S, Gorincioi G, Mura S, Pirtac M, Stancul I, Tcaciuc D, Albert M, Alexopoulou I, Arnaout A, Bartlett J, Engel J, Gilbert S, Parfitt J, Sekhon H, Thomas G, Rassl DM, Rintoul RC, Bifulco C, Tamakawa R, Urba W, Hayward N, Timmers H, Antenucci A, Facciolo F, Grazi G, Marino M, Merola R, de Krijger R, Gimenez-Roqueplo AP, Piché A, Chevalier S, McKercher G, Birsoy K, Barnett G, Brewer C, Farver C, Naska T, Pennell NA, Raymond D, Schilero C, Smolenski K, Williams F, Morrison C, Borgia JA, Liptay MJ, Pool M, Seder CW, Junker K, Omberg L, Dinkin M, Manikhas G, Alvaro D, Bragazzi MC, Cardinale V, Carpino G, Gaudio E, Chesla D, Cottingham S, Dubina M, Moiseenko F, Dhanasekaran R, Becker KF, Janssen KP, Slotta-Huspenina J, Abdel-Rahman MH, Aziz D, Bell S, Cebulla CM, Davis A, Duell R, Elder JB, Hilty J, Kumar B, Lang J, Lehman NL, Mandt R, Nguyen P, Pilarski R, Rai K, Schoenfield L, Senecal K, Wakely P, Hansen P, Lechan R, Powers J, Tischler A, Grizzle WE, Sexton KC, Kastl A, Henderson J, Porten S, Waldmann J, Fassnacht M, Asa SL, Schadendorf D, Couce M, Graefen M, Huland H, Sauter G, Schlomm T, Simon R, Tennstedt P, Olabode O, Nelson M, Bathe O, Carroll PR, Chan JM, Disaia P, Glenn P, Kelley RK, Landen CN, Phillips J, Prados M, Simko J, Smith-McCune K, VandenBerg S, Roggin K, Fehrenbach A, Kendler A, Sifri S, Steele R, Jimeno A, Carey F, Forgie I, Mannelli M, Carney M, Hernandez B, Campos B, Herold-Mende C, Jungk C, Unterberg A, von Deimling A, Bossler A, Galbraith J, Jacobus L, Knudson M, Knutson T, Ma D, Milhem M, Sigmund R, Godwin AK, Madan R, Rosenthal HG, Adebamowo C, Adebamowo SN, Boussioutas A, Beer D, Giordano T, Mes-Masson AM, Saad F, Bocklage T, Landrum L, Mannel R, Moore K, Moxley K, Postier R, Walker J, Zuna R, Feldman M, Valdivieso F, Dhir R, Luketich J, Pinero EMM, Quintero-Aguilo M, Carlotti CG Jr, Dos Santos JS, Kemp R, Sankarankuty A, Tirapelli D, Catto J, Agnew K, Swisher E, Creaney J, Robinson B, Shelley CS, Godwin EM, Kendall S, Shipman C, Bradford C, Carey T, Haddad A, Moyer J, Peterson L, Prince M, Rozek L, Wolf G, Bowman R, Fong KM, Yang I, Korst R, Rathmell WK, Fantacone-Campbell JL, Hooke JA, Kovatich AJ, Shriver CD, DiPersio J, Drake B, Govindan R, Heath S, Ley T, Van Tine B, Westervelt P, Rubin MA, Lee JI, Aredes ND, Mariamidze A. Author information: (1)Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. (2)Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Key Laboratory of Genomic and Precision Medicine, Gastrointestinal Cancer Research Center, Beijing Institute of Genomics, Chinese Academy of Sciences, 100101 Beijing, China. (3)Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. (4)Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. Electronic address: [email protected]. The role of enhancers, a key class of non-coding regulatory DNA elements, in cancer development has increasingly been appreciated. Here, we present the detection and characterization of a large number of expressed enhancers in a genome-wide analysis of 8928 tumor samples across 33 cancer types using TCGA RNA-seq data. Compared with matched normal tissues, global enhancer activation was observed in most cancers. Across cancer types, global enhancer activity was positively associated with aneuploidy, but not mutation load, suggesting a hypothesis centered on "chromatin-state" to explain their interplay. Integrating eQTL, mRNA co-expression, and Hi-C data analysis, we developed a computational method to infer causal enhancer-gene interactions, revealing enhancers of clinically actionable genes. Having identified an enhancer ∼140 kb downstream of PD-L1, a major immunotherapy target, we validated it experimentally. This study provides a systematic view of enhancer activity in diverse tumor contexts and suggests the clinical implications of enhancers. Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.cell.2018.03.027 PMCID: PMC5890960 PMID: 29625054 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34361106
1. Int J Mol Sci. 2021 Aug 3;22(15):8337. doi: 10.3390/ijms22158337. Regulation Network of Colorectal-Cancer-Specific Enhancers in the Progression of Colorectal Cancer. Chen B(1), Ma Y(1), Bi J(1), Wang W(1), He A(1), Su G(1), Zhao Z(1), Shi J(1), Zhang L(1). Author information: (1)State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China. Enhancers regulate multiple genes via higher-order chromatin structures, and they further affect cancer progression. Epigenetic changes in cancer cells activate several cancer-specific enhancers that are silenced in normal cells. These cancer-specific enhancers are potential therapeutic targets of cancer. However, the functions and regulation networks of colorectal-cancer-specific enhancers are still unknown. In this study, we profile colorectal-cancer-specific enhancers and reveal their regulation network through the analysis of HiChIP data that were derived from a colorectal cancer cell line and Hi-C and RNA-seq data that were derived from tissue samples by in silico analysis and in vitro experiments. Enhancer-promoter loops in colorectal cancer cells containing colorectal-cancer-specific enhancers are involved in more than 50% of the topological associated domains (TADs) changed in colorectal cancer cells compared to normal colon cells. In addition, colorectal-cancer-specific enhancers interact with 152 genes that are significantly and highly expressed in colorectal cancer cells. These colorectal-cancer-specific enhancer target genes include ITGB4, RECQL4, MSLN, and GDF15. We propose that the regulation network of colorectal-cancer-specific enhancers plays an important role in the progression of colorectal cancer. DOI: 10.3390/ijms22158337 PMCID: PMC8348541 PMID: 34361106 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35869079
1. Nat Commun. 2022 Jul 22;13(1):4247. doi: 10.1038/s41467-022-31919-8. A multi-omic dissection of super-enhancer driven oncogenic gene expression programs in ovarian cancer. Kelly MR(#)(1)(2), Wisniewska K(#)(1), Regner MJ(1)(2), Lewis MW(1), Perreault AA(3), Davis ES(2), Phanstiel DH(1)(3)(4), Parker JS(1)(2)(5), Franco HL(6)(7)(8). Author information: (1)Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. (2)Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. (3)Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. (4)Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. (5)Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. (6)Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. [email protected]. (7)Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. [email protected]. (8)Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. [email protected]. (#)Contributed equally The human genome contains regulatory elements, such as enhancers, that are often rewired by cancer cells for the activation of genes that promote tumorigenesis and resistance to therapy. This is especially true for cancers that have little or no known driver mutations within protein coding genes, such as ovarian cancer. Herein, we utilize an integrated set of genomic and epigenomic datasets to identify clinically relevant super-enhancers that are preferentially amplified in ovarian cancer patients. We systematically probe the top 86 super-enhancers, using CRISPR-interference and CRISPR-deletion assays coupled to RNA-sequencing, to nominate two salient super-enhancers that drive proliferation and migration of cancer cells. Utilizing Hi-C, we construct chromatin interaction maps that enable the annotation of direct target genes for these super-enhancers and confirm their activity specifically within the cancer cell compartment of human tumors using single-cell genomics data. Together, our multi-omic approach examines a number of fundamental questions about how regulatory information encoded into super-enhancers drives gene expression networks that underlie the biology of ovarian cancer. © 2022. The Author(s). DOI: 10.1038/s41467-022-31919-8 PMCID: PMC9307778 PMID: 35869079 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/33502116
1. Mol Syst Biol. 2021 Jan;17(1):e9873. doi: 10.15252/msb.20209873. Transcriptionally active enhancers in human cancer cells. Lidschreiber K(1)(2), Jung LA(2)(3), von der Emde H(1), Dave K(4), Taipale J(4)(5)(6), Cramer P(1)(2), Lidschreiber M(1)(2). Author information: (1)Department of Molecular Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany. (2)Department of Biosciences and Nutrition, Karolinska Institutet, NEO, Huddinge, Sweden. (3)Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Solna, Sweden. (4)Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Solna, Sweden. (5)Department of Biochemistry, University of Cambridge, Cambridge, UK. (6)Genome-Scale Biology Program, University of Helsinki, Helsinki, Finland. The growth of human cancer cells is driven by aberrant enhancer and gene transcription activity. Here, we use transient transcriptome sequencing (TT-seq) to map thousands of transcriptionally active putative enhancers in fourteen human cancer cell lines covering seven types of cancer. These enhancers were associated with cell type-specific gene expression, enriched for genetic variants that predispose to cancer, and included functionally verified enhancers. Enhancer-promoter (E-P) pairing by correlation of transcription activity revealed ~ 40,000 putative E-P pairs, which were depleted for housekeeping genes and enriched for transcription factors, cancer-associated genes, and 3D conformational proximity. The cell type specificity and transcription activity of target genes increased with the number of paired putative enhancers. Our results represent a rich resource for future studies of gene regulation by enhancers and their role in driving cancerous cell growth. © 2021 The Authors. Published under the terms of the CC BY 4.0 license. DOI: 10.15252/msb.20209873 PMCID: PMC7838827 PMID: 33502116 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/30867030
1. Clin Epigenetics. 2019 Mar 12;11(1):48. doi: 10.1186/s13148-019-0645-x. The hyper-activation of transcriptional enhancers in breast cancer. Li QL(1)(2), Wang DY(1), Ju LG(3), Yao J(1)(2), Gao C(1)(2), Lei PJ(1)(2), Li LY(1)(2), Zhao XL(4), Wu M(5)(6). Author information: (1)Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China. (2)Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Enteropathy, Wuhan University, Wuhan, 430072, Hubei, China. (3)Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, 430072, Hubei, China. (4)Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China. [email protected]. (5)Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, China. [email protected]. (6)Hubei Key Laboratory of Developmentally Originated Disease, Hubei Key Laboratory of Enteropathy, Wuhan University, Wuhan, 430072, Hubei, China. [email protected]. BACKGROUND: Activation of transcription enhancers, especially super-enhancers, is one of the critical epigenetic features of tumorigenesis. However, very few studies have systematically identified the enhancers specific in cancer tissues. METHODS: Here, we studied the change of histone modifications in MMTV-PyVT breast cancer model, combining mass spectrometry-based proteomics and ChIP-seq-based epigenomics approaches. Some of the proteomic results were confirmed with western blotting and IHC staining. An inhibitor of H3K27ac was applied to study its effect on cancer development. RESULTS: H3K27ac and H4K8ac are elevated in cancer, which was confirmed in patient tissue chips. ChIP-seq revealed that H4K8ac is co-localized with H3K27ac on chromatin, especially on distal enhancers. Epigenomic studies further identified a subgroup of super-enhancers marked by H3K4me3 peaks in the intergenic regions. The H3K4me3-enriched regions enhancers are associated with higher level of H3K27ac and H4K8ac compared with the average level of conventional super-enhancers and are associated with higher transcription level of their adjacent genes. We identified 148 H3K4me3-enriched super-enhancers with higher gene expression in tumor, which may be critical for breast cancer. One inhibitor for p300 and H3K27ac, C646, repressed tumor formation probably through inhibiting Vegfa and other genes. CONCLUSIONS: Taken together, our work identifies novel regulators and provides important resource to the genome-wide enhancer studies in breast cancer and raises the possibility of cancer treatment through modulating enhancer activity. DOI: 10.1186/s13148-019-0645-x PMCID: PMC6417266 PMID: 30867030 [Indexed for MEDLINE] Conflict of interest statement: ETHICS APPROVAL AND CONSENT TO PARTICIPATE: All the animal operations were following the laboratory animal guidelines of Wuhan University and were approved by the Animal Experimentations Ethics Committee of Wuhan University (Protocol NO. 14110B). No patient study was involved and the consent to participate is not applicable. CONSENT FOR PUBLICATION: All the authors have read the manuscript and agreed to publish the manuscript. COMPETING INTERESTS: The authors declare that they have no competing interests. PUBLISHER’S NOTE: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
http://www.ncbi.nlm.nih.gov/pubmed/31874597
1. BMC Bioinformatics. 2019 Dec 24;20(Suppl 15):598. doi: 10.1186/s12859-019-3180-z. DEEPSEN: a convolutional neural network based method for super-enhancer prediction. Bu H(1), Hao J(1), Gan Y(2), Zhou S(3), Guan J(4). Author information: (1)Department of Computer Science and Technology, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China. (2)School of Computer Science and Technology, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China. (3)Shanghai Key Lab of Intelligent Information Processing, and School of Computer Science, Fudan University, 220 Handan Road, Shanghai, 200433, China. (4)Department of Computer Science and Technology, Tongji University, 4800 Cao'an Road, Shanghai, 201804, China. [email protected]. BACKGROUND: Super-enhancers (SEs) are clusters of transcriptional active enhancers, which dictate the expression of genes defining cell identity and play an important role in the development and progression of tumors and other diseases. Many key cancer oncogenes are driven by super-enhancers, and the mutations associated with common diseases such as Alzheimer's disease are significantly enriched with super-enhancers. Super-enhancers have shown great potential for the identification of key oncogenes and the discovery of disease-associated mutational sites. RESULTS: In this paper, we propose a new computational method called DEEPSEN for predicting super-enhancers based on convolutional neural network. The proposed method integrates 36 kinds of features. Compared with existing approaches, our method performs better and can be used for genome-wide prediction of super-enhancers. Besides, we screen important features for predicting super-enhancers. CONCLUSION: Convolutional neural network is effective in boosting the performance of super-enhancer prediction. DOI: 10.1186/s12859-019-3180-z PMCID: PMC6929276 PMID: 31874597 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/24656127
1. Mol Cell. 2014 Mar 20;53(6):859-66. doi: 10.1016/j.molcel.2014.02.033. Enhancer malfunction in cancer. Herz HM(1), Hu D(1), Shilatifard A(2). Author information: (1)Stowers Institute for Medical Research, Kansas City, MO 64110, USA. (2)Stowers Institute for Medical Research, Kansas City, MO 64110, USA. Electronic address: [email protected]. Why certain point mutations in a general transcription factor are associated with specific forms of cancer has been a major question in cancer biology. Enhancers are DNA regulatory elements that are key regulators of tissue-specific gene expression. Recent studies suggest that enhancer malfunction through point mutations in either regulatory elements or factors modulating enhancer-promoter communication could be the cause of tissue-specific cancer development. In this Perspective, we will discuss recent findings in the identification of cancer-related enhancer mutations and the role of Drosophila Trr and its human homologs, the MLL3 and MLL4/COMPASS-like complexes, as enhancer histone H3 lysine 4 (H3K4) monomethyltransferases functioning in enhancer-promoter communication. Recent genome-wide studies in the cataloging of somatic mutations in cancer have identified mutations in intergenic sequences encoding regulatory elements-and in MLL3 and MLL4 in both hematological malignancies and solid tumors. We propose that cancer-associated mutations in MLL3 and MLL4 exert their properties through the malfunction of Trr/MLL3/MLL4-dependent enhancers. Copyright © 2014 Elsevier Inc. All rights reserved. DOI: 10.1016/j.molcel.2014.02.033 PMCID: PMC4049186 PMID: 24656127 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26356674
1. Oncotarget. 2015 Oct 20;6(32):32509-25. doi: 10.18632/oncotarget.5085. The search for cis-regulatory driver mutations in cancer genomes. Poulos RC(1), Sloane MA(1), Hesson LB(1), Wong JW(1). Author information: (1)Prince of Wales Clinical School and Lowy Cancer Research Centre, UNSW Australia, Sydney, Australia. With the advent of high-throughput and relatively inexpensive whole-genome sequencing technology, the focus of cancer research has begun to shift toward analyses of somatic mutations in non-coding cis-regulatory elements of the cancer genome. Cis-regulatory elements play an important role in gene regulation, with mutations in these elements potentially resulting in changes to the expression of linked genes. The recent discoveries of recurrent TERT promoter mutations in melanoma, and recurrent mutations that create a super-enhancer regulating TAL1 expression in T-cell acute lymphoblastic leukaemia (T-ALL), have sparked significant interest in the search for other somatic cis-regulatory mutations driving cancer development. In this review, we look more closely at the TERT promoter and TAL1 enhancer alterations and use these examples to ask whether other cis-regulatory mutations may play a role in cancer susceptibility. In doing so, we make observations from the data emerging from recent research in this field, and describe the experimental and analytical approaches which could be adopted in the hope of better uncovering the true functional significance of somatic cis-regulatory mutations in cancer. DOI: 10.18632/oncotarget.5085 PMCID: PMC4741709 PMID: 26356674 [Indexed for MEDLINE] Conflict of interest statement: CONFLICTS OF INTEREST The authors declare no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32128448
1. NPJ Precis Oncol. 2020 Feb 12;4:2. doi: 10.1038/s41698-020-0108-z. eCollection 2020. Super-enhancer function and its application in cancer targeted therapy. Tang F(#)(1), Yang Z(#)(2), Tan Y(1), Li Y(1). Author information: (1)1Department of Clinical Laboratory, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 410013 Changsha, China. (2)2Department of Clinical Laboratory, Zhuhai People's Hospital & Zhuhai Hospital of Jinan University, 519000 Zhuhai, China. (#)Contributed equally Recently, super-enhancers (SEs) have been identified as a unique type of transcriptional regulation involved in cancer development. SEs exhibit a size, high transcription factor density, and strong binding to the transcriptional machinery compared with typical enhancers. SEs play an essential role in cell growth, differentiation, and disease initiation and progression including tumorigenesis. In particular, cancer-specific SEs have been proven to be key oncogenic drivers types of tumor cells. Furthermore, it has been confirmed that cancer-specific SEs can mediate the dysregulation of signaling pathways and promote cancer cell growth. Additionally, therapeutic strategies directly targeting SE components, for example, by disrupting SE structure or inhibiting SE cofactors, have shown a good curative effect on various cancers. © The Author(s) 2020. DOI: 10.1038/s41698-020-0108-z PMCID: PMC7016125 PMID: 32128448 Conflict of interest statement: Competing interestsThe authors declare no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/35437236
1. Yi Chuan. 2022 Apr 20;44(4):275-288. doi: 10.16288/j.yczz.21-440. The regulatory mechanisms by enhancers during cancer initiation and progression. Qi SH(1)(2), Wang QL(1)(2), Zhang JY(1)(2), Liu Q(1)(2), Li CY(1)(2)(3)(4). Author information: (1)School of Engineering Medicine, Beihang University, Beijing 100191, China. (2)School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China. (3)Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China. (4)Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China. Enhancer is a DNA sequence, and mainly acts in cis to regulate gene transcription. Due to the uncertainty in both location and distance between enhancers and their target genes, it is more complex and difficult to study the underlying regulatory mechanism of enhancers. Accumulating evidences indicate that enhancers are closely associated with the occurrence and development of diseases, such as cancer. Therefore, the studies of enhancers in cancer will be helpful to deeply unravel cancer pathogenesis and to promote the development of antitumor drugs. The related research is with great social significance and economic value. Currently, the identification of enhancers is insufficient. The regulatory mechanisms by enhancers during the initiation and progression of cancer and other diseases have not been fully delineated. In this review, we provide an overview of enhancers, super enhancers and their properties, followed by a description of enhancer prediction and identification at the genome-wide level. Finally, we summarize the regulatory roles of enhancers during diseases such as cancer in recent years, thereby providing a reference for the future exploration on enhancer regulatory mechanisms as well as cancer diagnosis and treatment. Publisher: 增强子是一段具有转录调控功能的DNA序列,主要通过顺式调控方式发挥作用。由于增强子及其调控基因在位置和距离上的不确定性,大大增加了研究增强子作用机制的复杂性和困难性。越来越多的证据表明,增强子与癌症等疾病的发生发展密切相关,因此开展癌症相关增强子的研究,将有助于全面解析癌症发病机制,并推动抗肿瘤药物的高效研发,具有重要的社会意义和经济价值。目前对于增强子的鉴定不充分,增强子在癌症和其他疾病中的发生发展调控机制尚未得到完整的解析。本文主要对增强子和超级增强子及其特性进行介绍,并在全基因组水平上对增强子的预测和鉴定进行了描述,最后总结了近年来增强子在癌症等疾病发生过程中所发挥的调控作用,从而为未来解析增强子调控机制以及癌症的诊断和治疗提供参考。. DOI: 10.16288/j.yczz.21-440 PMID: 35437236 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33858773
1. Trends Immunol. 2021 May;42(5):418-431. doi: 10.1016/j.it.2021.03.005. Epub 2021 Apr 12. Deregulation of enhancer structure, function, and dynamics in acute lymphoblastic leukemia. Belver L(1), Albero R(2), Ferrando AA(3). Author information: (1)Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA; Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, 08916, Spain. (2)Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA. (3)Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA; Department of Systems Biology, Columbia University, New York, NY, 10032, USA; Department of Pediatrics, Columbia University Medical Center, New York, NY, 10032, USA; Department of Pathology, Columbia University Medical Center, New York, NY, 10032, USA. Electronic address: [email protected]. Enhancers control dynamic changes in gene expression and orchestrate the tightly controlled transcriptional circuitries that direct and coordinate cell growth, proliferation, survival, lineage commitment, and differentiation during lymphoid development. Enhancer hijacking and neoenhancer formation at oncogene loci, as well as aberrant activation of oncogene-associated enhancers, can induce constitutive activation of self-perpetuating oncogenic transcriptional circuitries, and contribute to the malignant transformation of immature lymphoid progenitors in acute lymphoblastic leukemia (ALL). In this review, we present recent discoveries of the role of enhancer dynamics in mouse and human lymphoid development, and discuss how genetic and epigenetic alterations of enhancer function can promote leukemogenesis, and potential strategies for targeting the enhancer machinery in the treatment of ALL. Copyright © 2021 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.it.2021.03.005 PMCID: PMC8091164 PMID: 33858773 [Indexed for MEDLINE] Conflict of interest statement: Declaration of interests None declared by authors.
http://www.ncbi.nlm.nih.gov/pubmed/29691590
1. Cell Mol Life Sci. 2018 Jul;75(14):2537-2555. doi: 10.1007/s00018-018-2820-1. Epub 2018 Apr 24. Enhancer reprogramming in tumor progression: a new route towards cancer cell plasticity. Fagnocchi L(1), Poli V(2), Zippo A(3)(4)(5). Author information: (1)Laboratory of Chromatin Biology and Epigenetics, Center for Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy. [email protected]. (2)Laboratory of Chromatin Biology and Epigenetics, Center for Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy. (3)Laboratory of Chromatin Biology and Epigenetics, Center for Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy. [email protected]. (4)Department of Epigenetics, Fondazione Istituto Nazionale di Genetica Molecolare "Romeo ed Enrica Invernizzi", Via F. Sforza 35, 20122, Milan, Italy. [email protected]. (5)Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy. [email protected]. Cancer heterogeneity arises during tumor progression as a consequence of genetic insults, environmental cues, and reversible changes in the epigenetic state, favoring tumor cell plasticity. The role of enhancer reprogramming is emerging as a relevant field in cancer biology as it supports adaptation of cancer cells to those environmental changes encountered during tumor progression and metastasis seeding. In this review, we describe the cancer-related alterations that drive oncogenic enhancer activity, leading to dysregulated transcriptional programs. We discuss the molecular mechanisms of both cis- and trans-factors in overriding the regulatory circuits that maintain cell-type specificity and imposing an alternative, de-regulated enhancer activity in cancer cells. We further comment on the increasing evidence which implicates stress response and aging-signaling pathways in the enhancer landscape reprogramming during tumorigenesis. Finally, we focus on the potential therapeutic implications of these enhancer-mediated subverted transcriptional programs, putting particular emphasis on the lack of information regarding tumor progression and the metastatic outgrowth, which still remain the major cause of mortality related to cancer. DOI: 10.1007/s00018-018-2820-1 PMCID: PMC11105402 PMID: 29691590 [Indexed for MEDLINE] Conflict of interest statement: The authors declare that they have no conflict of interests.
http://www.ncbi.nlm.nih.gov/pubmed/36250332
1. BMJ Open. 2022 Jul 19;12(7):e061841. doi: 10.1136/bmjopen-2022-061841. Multicentre, randomised controlled trial to investigate the effects of parental touch on relieving acute procedural pain in neonates (Petal). Cobo MM(1)(2), Moultrie F(1), Hauck AGV(1), Crankshaw D(1), Monk V(1), Hartley C(1), Evans Fry R(1), Robinson S(1), van der Vaart M(1), Baxter L(1), Adams E(3), Poorun R(4)(5), Bhatt A(1), Slater R(6). Author information: (1)Department of Paediatrics, University of Oxford, Oxford, UK. (2)Colegio de Ciencias Biologicas y Ambientales, Universidad San Francisco de Quito USFQ, Quito, Ecuador. (3)Newborn Care Unit, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK. (4)Children's Services, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK. (5)College of Medicine & Health, University of Exeter, Exeter, UK. (6)Department of Paediatrics, University of Oxford, Oxford, UK [email protected]. INTRODUCTION: Newborn infants routinely undergo minor painful procedures as part of postnatal care, with infants born sick or premature requiring a greater number of procedures. As pain in early life can have long-term neurodevelopmental consequences and lead to parental anxiety and future avoidance of interventions, effective pain management is essential. Non-pharmacological comfort measures such as breastfeeding, swaddling and sweet solutions are inconsistently implemented and are not always practical or effective in reducing the transmission of noxious input to the brain. Stroking of the skin can activate C-tactile fibres and reduce pain, and therefore could provide a simple and safe parent-led intervention for the management of pain. The trial aim is to determine whether parental touch prior to a painful clinical procedure provides effective pain relief in neonates. METHODS AND ANALYSIS: This is a multicentre randomised controlled trial. A total of 112 neonates born at 35 weeks' gestation or more requiring a blood test in the first week of life will be recruited and randomised to receive parental stroking either preprocedure or postprocedure. We will record brain activity (EEG), cardiac and respiratory dynamics, oxygen saturation and facial expression to provide proxy pain outcome measures. The primary outcome will be the reduction of noxious-evoked brain activity in response to a heel lance. Secondary outcomes will be a reduction in clinical pain scores (Premature Infant Pain Profile-Revised), postprocedural tachycardia and parental anxiety. ETHICS AND DISSEMINATION: The study has been approved by the London-South East Research Ethics Committee (ref: 21/LO/0523). The results will be widely disseminated through peer-reviewed publications, international conferences and via our partner neonatal charities Bliss and Supporting the Sick Newborn And their Parents (SSNAP). If the parental tactile intervention is effective, recommendations will be submitted via the National Health Service clinical guideline adoption process. STUDY STATUS: Commenced September 2021. TRIAL REGISTRATION NUMBER: NCT04901611; 14 135 962. © Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY. Published by BMJ. DOI: 10.1136/bmjopen-2022-061841 PMCID: PMC9301810 PMID: 36250332 [Indexed for MEDLINE] Conflict of interest statement: Competing interests: None declared.
http://www.ncbi.nlm.nih.gov/pubmed/33664472
1. J Perinatol. 2021 Sep;41(9):2298-2303. doi: 10.1038/s41372-021-01025-6. Epub 2021 Mar 4. New perspective for pain control in neonates: a comparative effectiveness research. Bresesti I(1)(2), Vanzù G(1), Redaelli F(2), Daniele I(1), Zuccotti GV(2), Cerritelli F(3), Lista G(#)(4), Fabiano V(#)(2). Author information: (1)Division of Neonatology, "V. Buzzi" Children's Hospital, ASST-FBF-Sacco, Milan, Italy. (2)Department of Pediatrics, "V. Buzzi" Children's Hospital, University of Milan, Milan, Italy. (3)Clinical human-based research Department, Foundation COME collaboration, Pescara, Italy. (4)Division of Neonatology, "V. Buzzi" Children's Hospital, ASST-FBF-Sacco, Milan, Italy. [email protected]. (#)Contributed equally OBJECTIVE: To compare sucrose 24% oral gel formulation to liquid formula and breastfeeding during a heel prick in neonates. STUDY DESIGN: In this comparative effectiveness research 195 neonates >36 weeks' gestation were randomised to three groups, receiving during heel stick: (i) breastfeeding, (ii) sucrose 24% liquid with non-nutritive sucking and (iii) sucrose 24% gel with non-nutritive sucking. The pain was assessed through the Neonatal Infant Pain Scale. RESULTS: All the methods analysed has shown to be effective in reducing pain. There was an increase in odds of pain following liquid sucrose compared to breastfeeding (OR = 1.60; 95% CI: 0.82-3.3; p = 0.17). A reduction of odds of pain was showed comparing sucrose to breastfeeding (OR = 0.78; 0.38-1.6; 0.48), and comparing sucrose gel to liquid formula (OR = 0.48; 0.23-0.96; p = 0.04). CONCLUSION: Sucrose 24% gel with non-nutritive sucking seems to be a valid alternative when breastfeeding is not possible. Further research is needed. © 2021. The Author(s), under exclusive licence to Springer Nature America, Inc. part of Springer Nature. DOI: 10.1038/s41372-021-01025-6 PMID: 33664472 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31317627
1. J Paediatr Child Health. 2020 Feb;56(2):207-214. doi: 10.1111/jpc.14559. Epub 2019 Jul 17. Oral sucrose for analgesia in children aged between 3 months and 3 years undergoing transurethral bladder catheterisation: A randomised, double-blinded, clinical trial. London K(1), Watson H(1), Kwok S(2), Nanan R(2), Liu A(2). Author information: (1)Department of Nuclear Medicine, Children's Hospital at Westmead, Sydney, New South Wales, Australia. (2)Department of Paediatrics, Nepean Hospital, Sydney, New South Wales, Australia. AIM: Many children admitted to hospital undergo invasive, painful and stressful procedures, including children who are not toilet trained undergoing transurethral bladder catheterisation (TUBC). Oral sucrose is commonly given to children to reduce procedural pain. In this study, we evaluated the effectiveness of oral sucrose in reducing procedural pain in children aged between 3 months and 3 years undergoing TUBC. METHODS: This study was a randomised, double-blind, placebo-controlled study conducted at Nepean Hospital, Sydney, Australia from June 2005 to June 2010. A total of 40 participants requiring TUBC for diagnostic evaluation were included. The participants were randomly assigned to receive 4 mL of 75% oral sucrose (n = 20) or a placebo (sterilised water) (n = 20). The primary outcomes were changes in two paediatric pain scale scores (the FLACC pain scale and the OUCHER pain scale), assessed by the parent/guardian(s), the doctor performing the TUBC and the nurse assisting. The secondary outcomes were physiological (changes in heart rate) and behavioural pain (crying) indicators. RESULTS: Of the outcome measures, 65% favoured the oral sucrose group, 31% favoured the placebo group, and 4% found no difference between the oral sucrose and placebo groups. CONCLUSION: While the trends favouring the sucrose group in this study were encouraging, as the results were not statistically significant, there was insufficient evidence to demonstrate the effectiveness of oral sucrose in reducing procedural pain in children aged between 3 months and 3 years undergoing TUBC. © 2019 Paediatrics and Child Health Division (The Royal Australasian College of Physicians). DOI: 10.1111/jpc.14559 PMID: 31317627 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/12867081
1. Biol Cell. 2003 May-Jun;95(3-4):169-78. doi: 10.1016/s0248-4900(03)00033-9. Generation of protein isoform diversity by alternative initiation of translation at non-AUG codons. Touriol C(1), Bornes S, Bonnal S, Audigier S, Prats H, Prats AC, Vagner S. Author information: (1)Institut National de la Santé et de la Recherche Médicale U589, Hormones, Facteurs de Croissance et Physiopathologie Vasculaire, Institut Fédératif de Recherche Louis Bugnard, C.H.U. Rangueil, 31403 Toulouse cedex 04, France. The use of several translation initiation codons in a single mRNA, by expressing several proteins from a single gene, contributes to the generation of protein diversity. A small, yet growing, number of mammalian mRNAs initiate translation from a non-AUG codon, in addition to initiating at a downstream in-frame AUG codon. Translation initiation on such mRNAs results in the synthesis of proteins harbouring different amino terminal domains potentially conferring on these isoforms distinct functions. Use of non-AUG codons appears to be governed by several features, including the sequence context and the secondary structure surrounding the codon. Selection of the downstream initiation codon can occur by leaky scanning of the 43S ribosomal subunit, internal entry of ribosome or ribosomal shunting. The biological significance of non-AUG alternative initiation is demonstrated by the different subcellular localisations and/or distinct biological functions of the isoforms translated from the single mRNA as illustrated by the two main angiogenic factor genes encoding the fibroblast growth factor 2 (FGF2) and the vascular endothelial growth factor (VEGF). Consequently, the regulation of alternative initiation of translation might have a crucial role for the biological function of the gene product. DOI: 10.1016/s0248-4900(03)00033-9 PMID: 12867081 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/21266472
1. Nucleic Acids Res. 2011 May;39(10):4220-34. doi: 10.1093/nar/gkr007. Epub 2011 Jan 25. Identification of evolutionarily conserved non-AUG-initiated N-terminal extensions in human coding sequences. Ivanov IP(1), Firth AE, Michel AM, Atkins JF, Baranov PV. Author information: (1)BioSciences Institute, University College Cork, Cork, Ireland. [email protected] In eukaryotes, it is generally assumed that translation initiation occurs at the AUG codon closest to the messenger RNA 5' cap. However, in certain cases, initiation can occur at codons differing from AUG by a single nucleotide, especially the codons CUG, UUG, GUG, ACG, AUA and AUU. While non-AUG initiation has been experimentally verified for a handful of human genes, the full extent to which this phenomenon is utilized--both for increased coding capacity and potentially also for novel regulatory mechanisms--remains unclear. To address this issue, and hence to improve the quality of existing coding sequence annotations, we developed a methodology based on phylogenetic analysis of predicted 5' untranslated regions from orthologous genes. We use evolutionary signatures of protein-coding sequences as an indicator of translation initiation upstream of annotated coding sequences. Our search identified novel conserved potential non-AUG-initiated N-terminal extensions in 42 human genes including VANGL2, FGFR1, KCNN4, TRPV6, HDGF, CITED2, EIF4G3 and NTF3, and also affirmed the conservation of known non-AUG-initiated extensions in 17 other genes. In several instances, we have been able to obtain independent experimental evidence of the expression of non-AUG-initiated products from the previously published literature and ribosome profiling data. DOI: 10.1093/nar/gkr007 PMCID: PMC3105428 PMID: 21266472 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23748256
1. Pain Res Manag. 2013 May-Jun;18(3):153-61. doi: 10.1155/2013/956549. A systematic review and meta-analyses of nonsucrose sweet solutions for pain relief in neonates. Bueno M(1), Yamada J, Harrison D, Khan S, Ohlsson A, Adams-Webber T, Beyene J, Stevens B. Author information: (1)School of Nursing, Federal University of Minas Gerais, Belo Horizonte, Brazil. [email protected] BACKGROUND: Sucrose has been demonstrated to provide analgesia for minor painful procedures in infants. However, results of trials investigating other sweet solutions for neonatal pain relief have not yet been synthesized. OBJECTIVE: To establish the efficacy of nonsucrose sweet-tasting solutions for pain relief during painful procedures in neonates. METHOD: The present article is a systematic review and meta-analyses of the literature. Standard methods of the Cochrane Neonatal Collaborative Review Group were used. Literature searches were reviewed for randomized controlled trials investigating the use of sweet solutions, except sucrose, for procedural pain management in neonates. Outcomes assessed included validated pain measures and behavioural and physiological indicators. RESULTS: Thirty-eight studies (3785 neonates) were included, 35 of which investigated glucose. Heel lancing was performed in 21⁄38 studies and venipuncture in 11⁄38 studies. A 3.6-point reduction in Premature Infant Pain Profile scores during heel lances was observed in studies comparing 20% to 30% glucose with no intervention (two studies, 124 neonates; mean difference -3.6 [95% CI -4.6 to -2.6]; P<0.001; I2=54%). A significant reduction in the incidence of cry after venipuncture for infants receiving 25% to 30% glucose versus water or no intervention was observed (three studies, 130 infants; risk difference -0.18 [95% CI -0.31 to -0.05]; P=0.008, number needed to treat = 6 [95% CI 3 to 20]; I2=63%). CONCLUSIONS: The present systematic review and meta-analyses demonstrate that glucose reduces pain scores and crying during single heel lances and venipunctures. Results indicate that 20% to 30% glucose solutions have analgesic effects and can be recommended as an alternative to sucrose for procedural pain reduction in healthy term and preterm neonates. HISTORIQUE : Il est démontré que le saccharose sert d’analgésie lors d’interventions mineures douloureuses chez les nourrissons. Cependant, il n’existe pas de synthèse des résultats d’essais sur d’autres solutions sucrées pour soulager la douleur. OBJECTIFS : Établir l’efficacité de solutions au goût sucré sans saccharose pour soulager la douleur pendant des interventions douloureuses chez les nouveau-nés. MÉTHODOLOGIE : Le présent article se compose d’une analyse systématique et de méta-analyses des publications. Il fait appel aux méthodes standards du Cochrane Neonatal Collaborative Review Group. Les chercheurs ont analysé les recherches dans les publications pour trouver des essais aléatoires et contrôlés sur l’utilisation de solutions sucrées, à part le saccharose, pour gérer la douleur causée par des interventions chez les nouveau-nés. Les issues évaluées incluaient les mesures de douleur validées et les indicateurs comportementaux et physiologiques. RÉSULTATS : Trente-huit études (3 785 nouveau-nés) étaient incluses, dont 35 portaient sur le glucose. Des incisions au talon ont été exécutées dans 21 des 38 études et une veinopuncture, dans 11 des 38 études. On observait une diminution de 3,6 points aux indices du profil de douleur des nourrissons prématurés pendant l’incision au talon dans les études comparant de 20 % à 30 % de glucose à l’absence d’intervention (deux études, 124 nouveau-nés, différence moyenne de −3,6 [95 % IC −4,6 à −2,6]; P<0,001). On observait également une réduction significative de l’incidence de pleurs après la veinopuncture chez les nourrissons qui recevaient de 25 % à 30 % de glucose par rapport à de l’eau ou à l’absence d’intervention (trois études, 130 nourrissons; différence de risque de −0,18 [95 % IC −0,31 à −0,05]; P=0,008, nombre nécessaire pour traiter = 6 [95 % IC 3 à 20]; I2=63 %). CONCLUSIONS : La présente analyse systématique et les présentes méta-analyses ont démontré que le glucose réduit les indices de douleur et les pleurs pendant de simples incisions au talon et des veinopunctures. Selon les résultats, des solutions de 20 % à 30 % de glucose ont des effets analgésiques et peuvent être recommandées pour remplacer le saccharose en vue de réduire la douleur lors d’interventions chez les nouveau-nés en santé à terme et prématurés. DOI: 10.1155/2013/956549 PMCID: PMC3673933 PMID: 23748256 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28369664
1. Curr Protoc Protein Sci. 2017 Apr 3;88:6.14.1-6.14.3. doi: 10.1002/cpps.29. N-Terminal Methionine Processing. Wingfield PT(1). Author information: (1)Protein Expression Laboratory, NIAMS/NIH, Bethesda, Maryland. Protein synthesis is initiated by methionine in eukaryotes and by formylmethionine in prokaryotes. N-terminal methionine can be co-translationally cleaved by the enzyme methionine aminopeptidase (MAP). When recombinant proteins are expressed in bacterial and mammalian expression systems, there is a simple universal rule that predicts whether the initiating methionine will be processed by MAP based on the size of the residue adjacent (penultimate) to the N-methionine. In general, if the side chains of the penultimate residues have a radius of gyration of 1.29 Å or less, methionine is cleaved. © 2017 by John Wiley & Sons, Inc. Copyright © 2017 John Wiley & Sons, Inc. DOI: 10.1002/cpps.29 PMCID: PMC5663234 PMID: 28369664 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28334756
1. Nucleic Acids Res. 2017 Apr 20;45(7):3615-3626. doi: 10.1093/nar/gkx070. Measurements of translation initiation from all 64 codons in E. coli. Hecht A(1)(2)(3), Glasgow J(1)(2)(3), Jaschke PR(3)(4), Bawazer LA(1)(2)(3), Munson MS(1)(2)(3), Cochran JR(1)(3), Endy D(1)(3), Salit M(1)(2)(3). Author information: (1)Joint Initiative for Metrology in Biology, Stanford, CA 94305, USA. (2)Genome-scale Measurements Group, National Institute of Standards and Technology, Stanford, CA 94305, USA. (3)Department of Bioengineering, Stanford, CA 94305, USA. (4)Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia. Our understanding of translation underpins our capacity to engineer living systems. The canonical start codon (AUG) and a few near-cognates (GUG, UUG) are considered as the 'start codons' for translation initiation in Escherichia coli. Translation is typically not thought to initiate from the 61 remaining codons. Here, we quantified translation initiation of green fluorescent protein and nanoluciferase in E. coli from all 64 triplet codons and across a range of DNA copy number. We detected initiation of protein synthesis above measurement background for 47 codons. Translation from non-canonical start codons ranged from 0.007 to 3% relative to translation from AUG. Translation from 17 non-AUG codons exceeded the highest reported rates of non-cognate codon recognition. Translation initiation from non-canonical start codons may contribute to the synthesis of peptides in both natural and synthetic biological systems. Published by Oxford University Press on behalf of Nucleic Acids Research 2017. DOI: 10.1093/nar/gkx070 PMCID: PMC5397182 PMID: 28334756 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/15215523
1. Protein Sci. 2004 Jul;13(7):1802-10. doi: 10.1110/ps.04679104. Removal of N-terminal methionine from recombinant proteins by engineered E. coli methionine aminopeptidase. Liao YD(1), Jeng JC, Wang CF, Wang SC, Chang ST. Author information: (1)Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan 115. [email protected] The removal of N-terminal translation initiator Met by methionine aminopeptidase (MetAP) is often crucial for the function and stability of proteins. On the basis of crystal structure and sequence alignment of MetAPs, we have engineered Escherichia coli MetAP by the mutation of three residues, Y168G, M206T, Q233G, in the substrate-binding pocket. Our engineered MetAPs are able to remove the Met from bulky or acidic penultimate residues, such as Met, His, Asp, Asn, Glu, Gln, Leu, Ile, Tyr, and Trp, as well as from small residues. The penultimate residue, the second residue after Met, was further removed if the antepenultimate residue, the third residue after Met, was small. By the coexpression of engineered MetAP in E. coli through the same or a separate vector, we have successfully produced recombinant proteins possessing an innate N terminus, such as onconase, an antitumor ribonuclease from the frog Rana pipiens. The N-terminal pyroglutamate of recombinant onconase is critical for its structural integrity, catalytic activity, and cyto-toxicity. On the basis of N-terminal sequence information in the protein database, 85%-90% of recombinant proteins should be produced in authentic form by our engineered MetAPs. DOI: 10.1110/ps.04679104 PMCID: PMC2279930 PMID: 15215523 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/9710598
1. Mol Cell Biol. 1998 Sep;18(9):5140-7. doi: 10.1128/MCB.18.9.5140. Initiation of protein synthesis in mammalian cells with codons other than AUG and amino acids other than methionine. Drabkin HJ(1), RajBhandary UL. Author information: (1)Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA. Protein synthesis is initiated universally with the amino acid methionine. In Escherichia coli, studies with anticodon sequence mutants of the initiator methionine tRNA have shown that protein synthesis can be initiated with several other amino acids. In eukaryotic systems, however, a yeast initiator tRNA aminoacylated with isoleucine was found to be inactive in initiation in mammalian cell extracts. This finding raised the question of whether methionine is the only amino acid capable of initiation of protein synthesis in eukaryotes. In this work, we studied the activities, in initiation, of four different anticodon sequence mutants of human initiator tRNA in mammalian COS1 cells, using reporter genes carrying mutations in the initiation codon that are complementary to the tRNA anticodons. The mutant tRNAs used are aminoacylated with glutamine, methionine, and valine. Our results show that in the presence of the corresponding mutant initiator tRNAs, AGG and GUC can initiate protein synthesis in COS1 cells with methionine and valine, respectively. CAG initiates protein synthesis with glutamine but extremely poorly, whereas UAG could not be used to initiate protein synthesis with glutamine. We discuss the potential applications of the mutant initiator tRNA-dependent initiation of protein synthesis with codons other than AUG for studying the many interesting aspects of protein synthesis initiation in mammalian cells. DOI: 10.1128/MCB.18.9.5140 PMCID: PMC109099 PMID: 9710598 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/8199241
1. Biochimie. 1993;75(12):1061-75. doi: 10.1016/0300-9084(93)90005-d. Methionine as translation start signal: a review of the enzymes of the pathway in Escherichia coli. Meinnel T(1), Mechulam Y, Blanquet S. Author information: (1)Laboratoire de Biochimie, URA-CNRS no 240, Palaiseau, France. Methionine is the universal translation start but the first methionine is removed from most mature proteins. This review focuses on our present knowledge of the five enzymes sustaining the methionine pathway in translation initiation in Escherichia coli: methionyl-tRNA synthetase, methionyl-tRNA(fMet) formyltransferase, peptidyl-tRNA hydrolase, peptide deformylase and methionine aminopeptidase. The possible significance of retaining methionine as initiation signal is discussed. DOI: 10.1016/0300-9084(93)90005-d PMID: 8199241 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36173861
1. Science. 2022 Sep 30;377(6614):1533-1537. doi: 10.1126/science.abq5082. Epub 2022 Sep 29. Actin maturation requires the ACTMAP/C19orf54 protease. Haahr P(1)(2), Galli RA(3), van den Hengel LG(1)(4), Bleijerveld OB(5), Kazokaitė-Adomaitienė J(6), Song JY(7), Kroese LJ(8), Krimpenfort P(8), Baltissen MP(9), Vermeulen M(9), Ottenheijm CAC(3), Brummelkamp TR(1)(4). Author information: (1)Division of Biochemistry, Netherlands Cancer Institute, 1066CX Amsterdam, Netherlands. (2)Novo Nordisk Foundation Center for Protein Research (NNF-CPR), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark. (3)Department of Physiology, Amsterdam UMC (VUmc), 1081HV Amsterdam, Netherlands. (4)Oncode Institute, Division of Biochemistry, Netherlands Cancer Institute, 1066CX Amsterdam, Netherlands. (5)Proteomics Facility, Netherlands Cancer Institute, 1066CX Amsterdam, Netherlands. (6)Protein Facility, Division of Biochemistry, Netherlands Cancer Institute, 1066CX Amsterdam, Netherlands. (7)Animal Pathology, Netherlands Cancer Institute, 1066CX Amsterdam, Netherlands. (8)Animal Modeling Facility, Netherlands Cancer Institute, 1066CX Amsterdam, Netherlands. (9)Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525GA Nijmegen, Netherlands. Protein synthesis generally starts with a methionine that is removed during translation. However, cytoplasmic actin defies this rule because its synthesis involves noncanonical excision of the acetylated methionine by an unidentified enzyme after translation. Here, we identified C19orf54, named ACTMAP (actin maturation protease), as this enzyme. Its ablation resulted in viable mice in which the cytoskeleton was composed of immature actin molecules across all tissues. However, in skeletal muscle, the lengths of sarcomeric actin filaments were shorter, muscle function was decreased, and centralized nuclei, a common hallmark of myopathies, progressively accumulated. Thus, ACTMAP encodes the missing factor required for the synthesis of mature actin and regulates specific actin-dependent traits in vivo. DOI: 10.1126/science.abq5082 PMID: 36173861 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/11738381
1. Trends Plant Sci. 2001 Dec;6(12):566-72. doi: 10.1016/s1360-1385(01)02151-3. Organellar peptide deformylases: universality of the N-terminal methionine cleavage mechanism. Giglione C(1), Meinnel T. Author information: (1)Institut des Sciences du Végétal, UPR2355, Centre National de la Recherche Scientifique, Bâtiment 23, 1 avenue de la Terrasse, F-91198 Gif-sur-Yvette Cedex, France. [email protected] Most mature proteins do not retain their initial N-terminal amino acid (methionine in the cytosol and N-formyl methionine in the organelles). Recent studies have shown that dedicated machinery is involved in this process in plants. In addition to cytosolic and organelle-targeted methionine aminopeptidases, organellar peptide deformylases have been identified. Here, we attempt to answer questions about the mechanism, specificity and significance of N-terminal methionine cleavage in plant organelles. It seems to be universal because orthologues of plant deformylases are found in most living organisms. DOI: 10.1016/s1360-1385(01)02151-3 PMID: 11738381 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25803486
1. Elife. 2015 Mar 24;4:e06857. doi: 10.7554/eLife.06857. Transcription factor MITF and remodeller BRG1 define chromatin organisation at regulatory elements in melanoma cells. Laurette P(1), Strub T(1), Koludrovic D(1), Keime C(1), Le Gras S(1), Seberg H(2), Van Otterloo E(2), Imrichova H(3), Siddaway R(4), Aerts S(3), Cornell RA(2), Mengus G(1), Davidson I(1). Author information: (1)Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Strasbourg, France. (2)University of Iowa College of Medicine, Iowa City, United States. (3)Laboratory of Computational Biology, Center for Human Genetics, University of Leuven, Leuven, Belgium. (4)Arthur and Sonia Labatt Brain Tumor Research Centre, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Canada. Microphthalmia-associated transcription factor (MITF) is the master regulator of the melanocyte lineage. To understand how MITF regulates transcription, we used tandem affinity purification and mass spectrometry to define a comprehensive MITF interactome identifying novel cofactors involved in transcription, DNA replication and repair, and chromatin organisation. We show that MITF interacts with a PBAF chromatin remodelling complex comprising BRG1 and CHD7. BRG1 is essential for melanoma cell proliferation in vitro and for normal melanocyte development in vivo. MITF and SOX10 actively recruit BRG1 to a set of MITF-associated regulatory elements (MAREs) at active enhancers. Combinations of MITF, SOX10, TFAP2A, and YY1 bind between two BRG1-occupied nucleosomes thus defining both a signature of transcription factors essential for the melanocyte lineage and a specific chromatin organisation of the regulatory elements they occupy. BRG1 also regulates the dynamics of MITF genomic occupancy. MITF-BRG1 interplay thus plays an essential role in transcription regulation in melanoma. DOI: 10.7554/eLife.06857 PMCID: PMC4407272 PMID: 25803486 [Indexed for MEDLINE] Conflict of interest statement: ID: Reviewing editor, eLife. The other authors declare that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/21496633
1. Handb Clin Neurol. 2011;101:167-80. doi: 10.1016/B978-0-08-045031-5.00013-X. Facioscapulohumeral dystrophy and scapuloperoneal syndromes. Orrell RW(1). Author information: (1)University Department of Clinical Neurosciences, UCL Institute of Neurology, London, UK. Facioscapulohumeral dystrophy (FSHD) is the third most common muscular dystrophy. It is named for its characteristic involvement of the muscles of the face and upper arm. It is present worldwide, with a prevalence of around 4 per 100000 and an incidence of about 1 in 20000. Overall lifespan is not affected significantly. The scapuloperoneal syndrome is a rarer presentation that may cause some confusion. FSHD is an autosomal dominant condition. The molecular genetics of FSHD are complex, with current understanding focusing on epigenetic effects related to contraction-dependent (FSHD1) and contraction-independent (FSHD2) effects of a hypomethylated repeat sequence (D4Z4), in the presence of a specific 4qA161 phenotype. Molecular genetic diagnosis is available based on these findings, but with some complexities which may lead to false-negative results on routine laboratory investigation. No medication has been demonstrated to alter the clinical course of the disease significantly. A range of supportive measures may be applied. This chapter reviews the epidemiology, pathogenesis, genetics, clinical features, investigation, prognosis, and management of patients with FSHD and the scapuloperoneal syndrome. Copyright © 2011 Elsevier Inc. All rights reserved. DOI: 10.1016/B978-0-08-045031-5.00013-X PMID: 21496633 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/15277526
1. J Biol Chem. 2004 Oct 1;279(40):41911-7. doi: 10.1074/jbc.M404964200. Epub 2004 Jul 23. Microphthalmia transcription factor induces both retinal pigmented epithelium and neural crest melanocytes from neuroretina cells. Planque N(1), Raposo G, Leconte L, Anezo O, Martin P, Saule S. Author information: (1)UMR 146, Institut Curie Section de Recherche, Bātiment 110, Centre Universitaire, 91405 Orsay Cedex, France. Mitf encodes a basic helix-loop-helix transcription factor that plays an essential role in the differentiation of the retinal pigmented epithelium (RPE) and neural crest-derived melanocytes. As cells containing melanogenic enzymes (TRP2) are found in Mitf mouse mutants, it is not clear whether Mitf is a downstream factor or a master regulator of melanocyte differentiation. To further study the role of Mitf in committing cells to the melanocyte lineage, we express Mitf in the cultured quail neuroretina cells. This leads to the induction of two types of pigmented cells: neural crest-derived melanocytes, according to their dendritic morphology, physiology, and gene expression pattern are observed together with pigmented epithelial RPE-like cells. The expression of Mitf is lower in pigmented epithelial RPE-like cells than in neural crest-derived melanocytes. Accordingly, overexpression of Mitf in cultured quail RPE causes cells to develop into neural crest-like pigmented cells. Thus, Mitf is sufficient for the proper differentiation of crest-like pigmented cells from retinal cells and its expression level may determine the type of pigment cell induced. DOI: 10.1074/jbc.M404964200 PMID: 15277526 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28649789
1. Pigment Cell Melanoma Res. 2017 Sep;30(5):454-466. doi: 10.1111/pcmr.12611. Beyond MITF: Multiple transcription factors directly regulate the cellular phenotype in melanocytes and melanoma. Seberg HE(1), Van Otterloo E(2), Cornell RA(1)(3). Author information: (1)Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA. (2)SDM-Craniofacial Biology, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA. (3)Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA. MITF governs multiple steps in the development of melanocytes, including specification from neural crest, growth, survival, and terminal differentiation. In addition, the level of MITF activity determines the phenotype adopted by melanoma cells, whether invasive, proliferative, or differentiated. However, MITF does not act alone. Here, we review literature on the transcription factors that co-regulate MITF-dependent genes. ChIP-seq studies have indicated that the transcription factors SOX10, YY1, and TFAP2A co-occupy subsets of regulatory elements bound by MITF in melanocytes. Analyses at single loci also support roles for LEF1, RB1, IRF4, and PAX3 acting in combination with MITF, while sequence motif analyses suggest that additional transcription factors colocalize with MITF at many melanocyte-specific regulatory elements. However, the precise biochemical functions of each of these MITF collaborators and their contributions to gene expression remain to be elucidated. Analogous to the transcriptional networks in morphogen-patterned tissues during embryogenesis, we anticipate that the level of MITF activity is controlled not only by the concentration of activated MITF, but also by additional transcription factors that either quantitatively or qualitatively influence the expression of MITF-target genes. © 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd. DOI: 10.1111/pcmr.12611 PMCID: PMC5939569 PMID: 28649789 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/21258399
1. Oncogene. 2011 May 19;30(20):2319-32. doi: 10.1038/onc.2010.612. Epub 2011 Jan 24. Essential role of microphthalmia transcription factor for DNA replication, mitosis and genomic stability in melanoma. Strub T(1), Giuliano S, Ye T, Bonet C, Keime C, Kobi D, Le Gras S, Cormont M, Ballotti R, Bertolotto C, Davidson I. Author information: (1)Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, Illkirch, France. Comment in Oncogene. 2011 May 19;30(20):2304-6. doi: 10.1038/onc.2010.641. Malignant melanoma is an aggressive cancer known for its notorious resistance to most current therapies. The basic helix-loop-helix microphthalmia transcription factor (MITF) is the master regulator determining the identity and properties of the melanocyte lineage, and is regarded as a lineage-specific 'oncogene' that has a critical role in the pathogenesis of melanoma. MITF promotes melanoma cell proliferation, whereas sustained supression of MITF expression leads to senescence. By combining chromatin immunoprecipitation coupled to high throughput sequencing (ChIP-seq) and RNA sequencing analyses, we show that MITF directly regulates a set of genes required for DNA replication, repair and mitosis. Our results reveal how loss of MITF regulates mitotic fidelity, and through defective replication and repair induces DNA damage, ultimately ending in cellular senescence. These findings reveal a lineage-specific control of DNA replication and mitosis by MITF, providing new avenues for therapeutic intervention in melanoma. The identification of MITF-binding sites and gene-regulatory networks establish a framework for understanding oncogenic basic helix-loop-helix factors such as N-myc or TFE3 in other cancers. DOI: 10.1038/onc.2010.612 PMID: 21258399 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/12819038
1. Am J Pathol. 2003 Jul;163(1):333-43. doi: 10.1016/S0002-9440(10)63657-7. MLANA/MART1 and SILV/PMEL17/GP100 are transcriptionally regulated by MITF in melanocytes and melanoma. Du J(1), Miller AJ, Widlund HR, Horstmann MA, Ramaswamy S, Fisher DE. Author information: (1)Department of Pediatric Hematology/Oncology, Dana-Farber Cancer Institute and Children's Hospital, Harvard Medical School, Boston, USA. The clinically important melanoma diagnostic antibodies HMB-45, melan-A, and MITF (D5) recognize gene products of the melanocyte-lineage genes SILV/PMEL17/GP100, MLANA/MART1, and MITF, respectively. MITF encodes a transcription factor that is essential for normal melanocyte development and appears to regulate expression of several pigmentation genes. In this report, the possibility was examined that MITF might additionally regulate expression of the SILV and MLANA genes. Both genes contain conserved MITF consensus DNA sequences that were bound by MITF in vitro and in vivo, based on electrophoretic mobility shift assay and chromatin-immunoprecipitation. In addition, MITF regulated their promoter/enhancer regions in reporter assays, and up- or down-regulation of MITF produced corresponding modulation of endogenous SILV and MLANA in melanoma cells. Expression patterns were compared with these factors in a series of melanoma cell lines whose mutational status of the proto-oncogene BRAF was also known. SILV and MLANA expression correlated with MITF, while no clear correlation was seen relative to BRAF mutation. Finally, mRNA expression array analysis of primary human melanomas demonstrated a tight correlation in their expression levels in clinical tumor specimens. Collectively, this study links three important melanoma antigens into a common transcriptional pathway regulated by MITF. DOI: 10.1016/S0002-9440(10)63657-7 PMCID: PMC1868174 PMID: 12819038 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23935512
1. PLoS Genet. 2013;9(7):e1003644. doi: 10.1371/journal.pgen.1003644. Epub 2013 Jul 25. A dual role for SOX10 in the maintenance of the postnatal melanocyte lineage and the differentiation of melanocyte stem cell progenitors. Harris ML(1), Buac K, Shakhova O, Hakami RM, Wegner M, Sommer L, Pavan WJ. Author information: (1)Genetic Disease Research Branch, National Human Genome Institute, National Institutes of Health, Bethesda, Maryland, United States of America. During embryogenesis, the transcription factor, Sox10, drives the survival and differentiation of the melanocyte lineage. However, the role that Sox10 plays in postnatal melanocytes is not established. We show in vivo that melanocyte stem cells (McSCs) and more differentiated melanocytes express SOX10 but that McSCs remain undifferentiated. Sox10 knockout (Sox10(fl); Tg(Tyr::CreER)) results in loss of both McSCs and differentiated melanocytes, while overexpression of Sox10 (Tg(DctSox10)) causes premature differentiation and loss of McSCs, leading to hair graying. This suggests that levels of SOX10 are key to normal McSC function and Sox10 must be downregulated for McSC establishment and maintenance. We examined whether the mechanism of Tg(DctSox10) hair graying is through increased expression of Mitf, a target of SOX10, by asking if haploinsufficiency for Mitf (Mitf(vga9) ) can rescue hair graying in Tg(DctSox10) animals. Surprisingly, Mitf(vga9) does not mitigate but exacerbates Tg(DctSox10) hair graying suggesting that MITF participates in the negative regulation of Sox10 in McSCs. These observations demonstrate that while SOX10 is necessary to maintain the postnatal melanocyte lineage it is simultaneously prevented from driving differentiation in the McSCs. This data illustrates how tissue-specific stem cells can arise from lineage-specified precursors through the regulation of the very transcription factors important in defining that lineage. DOI: 10.1371/journal.pgen.1003644 PMCID: PMC3723529 PMID: 23935512 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/26206884
1. Hum Mol Genet. 2015 Oct 1;24(19):5433-50. doi: 10.1093/hmg/ddv267. Epub 2015 Jul 23. Genomic analysis reveals distinct mechanisms and functional classes of SOX10-regulated genes in melanocytes. Fufa TD(1), Harris ML(1), Watkins-Chow DE(1), Levy D(1), Gorkin DU(2), Gildea DE(3), Song L(4), Safi A(4), Crawford GE(4), Sviderskaya EV(5), Bennett DC(5), Mccallion AS(2), Loftus SK(1), Pavan WJ(6). Author information: (1)Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA. (2)McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA. (3)Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA. (4)Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA, Department of Pediatrics, Division of Molecular Genetics, Duke University, Durham, NC 27708, USA and. (5)Molecular Cell Sciences Research Centre, St George's, University of London, London SW17 0RE, UK. (6)Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA, [email protected]. SOX10 is required for melanocyte development and maintenance, and has been linked to melanoma initiation and progression. However, the molecular mechanisms by which SOX10 guides the appropriate gene expression programs necessary to promote the melanocyte lineage are not fully understood. Here we employ genetic and epigenomic analysis approaches to uncover novel genomic targets and previously unappreciated molecular roles of SOX10 in melanocytes. Through global analysis of SOX10-binding sites and epigenetic characteristics of chromatin states, we uncover an extensive catalog of SOX10 targets genome-wide. Our findings reveal that SOX10 predominantly engages 'open' chromatin regions and binds to distal regulatory elements, including novel and previously known melanocyte enhancers. Integrated chromatin occupancy and transcriptome analysis suggest a role for SOX10 in both transcriptional activation and repression to regulate functionally distinct classes of genes. We demonstrate that distinct epigenetic signatures and cis-regulatory sequence motifs predicted to bind putative co-regulatory transcription factors define SOX10-activated and SOX10-repressed target genes. Collectively, these findings uncover a central role of SOX10 as a global regulator of gene expression in the melanocyte lineage by targeting diverse regulatory pathways. Published by Oxford University Press 2015. This work is written by (a) US Government employee(s) and is in the public domain in the US. DOI: 10.1093/hmg/ddv267 PMCID: PMC4572067 PMID: 26206884 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/12789278
1. Oncogene. 2003 May 19;22(20):3035-41. doi: 10.1038/sj.onc.1206443. Microphthalamia-associated transcription factor: a critical regulator of pigment cell development and survival. Widlund HR(1), Fisher DE. Author information: (1)Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA. The microphthalamia-associated transcription factor (MITF) is an integral transcriptional regulator in melanocyte, the lineage from which melanoma cells originate. This basic-helix-loop-helix-leucine-zipper (bHLHzip) protein is critical for melanocyte cell-fate choice during commitment from pluripotent precursor cells in the neural crest. Its role in differentiation pathways has been highlighted by its potent transcriptional and lineage-specific regulation of the three major pigment enzymes: tyrosinase, Tyrp1, and Dct as well as other pigmentation factors. However, the cellular functions of MITF seem to be wider than differentiation and cell-fate pathways alone, since melanocytes and melanoma cells appear to require an expression of this factor. Here, we discuss the transcriptional networks in which MITF is thought to reside and describe signaling pathways in the cell which impinge on MITF. Accumulating evidence supports the notion that MITF is involved in survival pathways during normal development as well as during neoplastic growth of melanoma. DOI: 10.1038/sj.onc.1206443 PMID: 12789278 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17237008
1. Bull Cancer. 2007 Jan;94(1):81-92. [Malignant melanoma and the role of the paradoxal protein Microphthalmia transcription factor]. [Article in French] Denat L(1), Larue L. Author information: (1)Developmental Genetics of Melanocytes, UMR146 CNRS, Institut Curie, Bât. 110, 91405, Orsay Cedex. [email protected] Mitf protein is a transcription factor involved all along the life of pigmented cells. This protein is located in the center of multiple signaling pathways which control differentiation, morphology, proliferation and survival of the various cells of the melanocyte lineage: melanoblasts, melanocytes and melanoma. Mitf plays a major role in melanoblasts differentiation, by inducing the key enzyme of melanogenesis, tyrosinase, and its secondary enzymes, Tyrp1 and Dct. Mitf regulates morphology and migration of melanocytes, particularly by regulating cytoskeleton organization and cell-cell adhesion. Mitf plays a double role of inducer/repressor of cellular proliferation. This protein inhibits cell cycle progression and prevents non-proper cell division. In few cases, Mitf can also induce cell cycle. A minimal quantity/activity of Mitf is necessary for melanoblast survival. Essential protein of the melanocyte lineage, Mitf was proposed as diagnostic/pronostic marker for cutaneous melanoma. However, could we then consider MITF as the unique marker of such a cancer? PMID: 17237008 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17000761
1. Mol Cell Biol. 2006 Dec;26(23):8914-27. doi: 10.1128/MCB.02299-05. Epub 2006 Sep 25. The microphthalmia-associated transcription factor Mitf interacts with beta-catenin to determine target gene expression. Schepsky A(1), Bruser K, Gunnarsson GJ, Goodall J, Hallsson JH, Goding CR, Steingrimsson E, Hecht A. Author information: (1)Biochemistry and Molecular Biology, Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland. Commitment to the melanocyte lineage is characterized by the onset of expression of the microphthalmia-associated transcription factor (Mitf). This transcription factor plays a fundamental role in melanocyte development and maintenance and seems to be crucial for the survival of malignant melanocytes. Furthermore, Mitf has been shown to be involved in cell cycle regulation and to play important functions in self-renewal and maintenance of melanocyte stem cells. Although little is known about how Mitf regulates these various processes, one possibility is that Mitf interacts with other regulators. Here we show that Mitf can interact directly with beta-catenin, the key mediator of the canonical Wnt signaling pathway. The Wnt signaling pathway plays a critical role in melanocyte development and is intimately involved in triggering melanocyte stem cell proliferation. Significantly, constitutive activation of this pathway is a feature of a number of cancers including malignant melanoma. Here we show that Mitf can redirect beta-catenin transcriptional activity away from canonical Wnt signaling-regulated genes toward Mitf-specific target promoters to activate transcription. Thus, by a feedback mechanism, Mitf can diversify the output of canonical Wnt signaling to enhance the repertoire of genes regulated by beta-catenin. Our results reveal a novel mechanism by which Wnt signaling and beta-catenin activate gene expression, with significant implications for our understanding of both melanocyte development and melanoma. DOI: 10.1128/MCB.02299-05 PMCID: PMC1636837 PMID: 17000761 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/24769727
1. Cell Death Differ. 2014 Aug;21(8):1250-61. doi: 10.1038/cdd.2014.44. Epub 2014 Apr 25. Identification of a ZEB2-MITF-ZEB1 transcriptional network that controls melanogenesis and melanoma progression. Denecker G(1), Vandamme N(1), Akay O(1), Koludrovic D(2), Taminau J(1), Lemeire K(3), Gheldof A(1), De Craene B(1), Van Gele M(4), Brochez L(4), Udupi GM(5), Rafferty M(6), Balint B(6), Gallagher WM(5), Ghanem G(7), Huylebroeck D(8), Haigh J(9), van den Oord J(10), Larue L(11), Davidson I(2), Marine JC(12), Berx G(1). Author information: (1)1] Unit of Molecular and Cellular Oncology, Inflammation Research Center, VIB, 9052 Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium. (2)Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, INSERM, Université de Strasbourg, Illkirch, France. (3)Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium. (4)Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium. (5)1] UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College, Dublin 4, Ireland [2] OncoMark Limited, Nova UCD, Belfield Innovation Park, University College Dublin, Belfield, Dublin 4, Ireland. (6)OncoMark Limited, Nova UCD, Belfield Innovation Park, University College Dublin, Belfield, Dublin 4, Ireland. (7)Institute Jules Bordet, Brussels, Belgium. (8)1] Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium [2] Department of Cell Biology, Erasmus MC, 3015 GE Rotterdam, The Netherlands. (9)1] Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium [2] Vascular Cell Biology Unit, Department for Molecular Biomedical Research, VIB, Ghent, Belgium. (10)Department of Pathology, University Hospital Leuven, KU Leuven, Leuven, Belgium. (11)Curie Institute, Developmental Genetics of Melanocytes, Centre National de la Recherche Scientifique (CNRS) UMR3347, Institut National de la Santé et de la Recherche Médicale (INSERM) U1021, Orsay, France. (12)1] Center for the Biology of Disease, Laboratory for Molecular Cancer Biology, VIB, Leuven, Belgium [2] Center for Human Genetics, KU Leuven, Leuven, Belgium. Deregulation of signaling pathways that control differentiation, expansion and migration of neural crest-derived melanoblasts during normal development contributes also to melanoma progression and metastasis. Although several epithelial-to-mesenchymal (EMT) transcription factors, such as zinc finger E-box binding protein 1 (ZEB1) and ZEB2, have been implicated in neural crest cell biology, little is known about their role in melanocyte homeostasis and melanoma. Here we show that mice lacking Zeb2 in the melanocyte lineage exhibit a melanoblast migration defect and, unexpectedly, a severe melanocyte differentiation defect. Loss of Zeb2 in the melanocyte lineage results in a downregulation of the Microphthalmia-associated transcription factor (Mitf) and melanocyte differentiation markers concomitant with an upregulation of Zeb1. We identify a transcriptional signaling network in which the EMT transcription factor ZEB2 regulates MITF levels to control melanocyte differentiation. Moreover, our data are also relevant for human melanomagenesis as loss of ZEB2 expression is associated with reduced patient survival. DOI: 10.1038/cdd.2014.44 PMCID: PMC4085532 PMID: 24769727 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28263292
1. Lab Invest. 2017 Jun;97(6):649-656. doi: 10.1038/labinvest.2017.9. Epub 2017 Mar 6. The master role of microphthalmia-associated transcription factor in melanocyte and melanoma biology. Kawakami A(1), Fisher DE(1). Author information: (1)Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. Certain transcription factors have vital roles in lineage development, including specification of cell types and control of differentiation. Microphthalmia-associated transcription factor (MITF) is a key transcription factor for melanocyte development and differentiation. MITF regulates expression of numerous pigmentation genes to promote melanocyte differentiation, as well as fundamental genes for maintaining cell homeostasis, including genes encoding proteins involved in apoptosis (eg, BCL2) and the cell cycle (eg, CDK2). Loss-of-function mutations of MITF cause Waardenburg syndrome type IIA, whose phenotypes include depigmentation due to melanocyte loss, whereas amplification or specific mutation of MITF can be an oncogenic event that is seen in a subset of familial or sporadic melanomas. In this article, we review basic features of MITF biological function and highlight key unresolved questions regarding this remarkable transcription factor. DOI: 10.1038/labinvest.2017.9 PMID: 28263292 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28249010
1. PLoS Genet. 2017 Mar 1;13(3):e1006636. doi: 10.1371/journal.pgen.1006636. eCollection 2017 Mar. TFAP2 paralogs regulate melanocyte differentiation in parallel with MITF. Seberg HE(1), Van Otterloo E(2), Loftus SK(3), Liu H(4), Bonde G(4), Sompallae R(5), Gildea DE(6), Santana JF(7), Manak JR(1)(7), Pavan WJ(3), Williams T(2), Cornell RA(1)(4). Author information: (1)Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America. (2)SDM-Craniofacial Biology, University of Colorado - Anschutz Medical Campus, Aurora, Colorado, United States of America. (3)Genetic Disease Research Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America. (4)Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States of America. (5)Bioinformatics Division, Iowa Institute of Human Genetics, University of Iowa, Iowa City, Iowa, United States of America. (6)Bioinformatics and Scientific Programming Core, Computational and Statistical Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America. (7)Department of Biology, University of Iowa, Iowa City, Iowa, United States of America. Comment in Pigment Cell Melanoma Res. 2017 Sep;30(5):449-451. doi: 10.1111/pcmr.12604. Mutations in the gene encoding transcription factor TFAP2A result in pigmentation anomalies in model organisms and premature hair graying in humans. However, the pleiotropic functions of TFAP2A and its redundantly-acting paralogs have made the precise contribution of TFAP2-type activity to melanocyte differentiation unclear. Defining this contribution may help to explain why TFAP2A expression is reduced in advanced-stage melanoma compared to benign nevi. To identify genes with TFAP2A-dependent expression in melanocytes, we profile zebrafish tissue and mouse melanocytes deficient in Tfap2a, and find that expression of a small subset of genes underlying pigmentation phenotypes is TFAP2A-dependent, including Dct, Mc1r, Mlph, and Pmel. We then conduct TFAP2A ChIP-seq in mouse and human melanocytes and find that a much larger subset of pigmentation genes is associated with active regulatory elements bound by TFAP2A. These elements are also frequently bound by MITF, which is considered the "master regulator" of melanocyte development. For example, the promoter of TRPM1 is bound by both TFAP2A and MITF, and we show that the activity of a minimal TRPM1 promoter is lost upon deletion of the TFAP2A binding sites. However, the expression of Trpm1 is not TFAP2A-dependent, implying that additional TFAP2 paralogs function redundantly to drive melanocyte differentiation, which is consistent with previous results from zebrafish. Paralogs Tfap2a and Tfap2b are both expressed in mouse melanocytes, and we show that mouse embryos with Wnt1-Cre-mediated deletion of Tfap2a and Tfap2b in the neural crest almost completely lack melanocytes but retain neural crest-derived sensory ganglia. These results suggest that TFAP2 paralogs, like MITF, are also necessary for induction of the melanocyte lineage. Finally, we observe a genetic interaction between tfap2a and mitfa in zebrafish, but find that artificially elevating expression of tfap2a does not increase levels of melanin in mitfa hypomorphic or loss-of-function mutants. Collectively, these results show that TFAP2 paralogs, operating alongside lineage-specific transcription factors such as MITF, directly regulate effectors of terminal differentiation in melanocytes. In addition, they suggest that TFAP2A activity, like MITF activity, has the potential to modulate the phenotype of melanoma cells. DOI: 10.1371/journal.pgen.1006636 PMCID: PMC5352137 PMID: 28249010 [Indexed for MEDLINE] Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/11830592
1. J Biol Chem. 2002 Apr 26;277(17):15132-41. doi: 10.1074/jbc.M200004200. Epub 2002 Feb 5. Pax3 down-regulation and shut-off of melanogenesis in melanoma B16/F10.9 by interleukin-6 receptor signaling. Kamaraju AK(1), Bertolotto C, Chebath J, Revel M. Author information: (1)Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel. The microphthalmia-associated transcription factor (Mitf) is essential for melanocytic lineage development and for expression of melanogenic enzymes, such as tyrosinase. Interleukin-6 receptor/interleukin-6 chimera (IL6RIL6) induces in B16/F10.9 melanoma cells a loss of melanogenesis preceded by a sharp decrease in Mitf mRNA and gene promoter activity. In the Mitf promoter, the main cis-acting element mediating the IL6RIL6 effect is shown to be the binding site of Pax3, a paired homeodomain factor regulating among other things the development of melanocytes. Pax3 protein and mRNA levels decline steadily after IL6RIL6 treatment, and overexpression of an ectopic Pax3 cDNA suppresses the Mitf promoter inhibition. Loss of the synergism between Pax3 and Sox10, a high mobility group domain costimulatory factor, seems to be critical in the rapid decrease in Mitf gene expression. The Pax3 down-regulation in IL6RIL6-induced F10.9 cell is linked to growth arrest and transdifferentiation to a glial cell phenotype. IL6RIL6 stimulates the interleukin-6 family cytokine receptor gp130, leading to the rapid phosphorylation of Stat3 on tyrosine 705. This phosphorylation is required for Pax3 down-regulation and Mitf promoter silencing since these are inhibited in F10.9 cells overexpressing the Stat3 DN-mutant Y705F. DOI: 10.1074/jbc.M200004200 PMID: 11830592 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/10770922
1. J Biol Chem. 2000 Jul 21;275(29):21920-7. doi: 10.1074/jbc.M000035200. The gene encoding the T-box factor Tbx2 is a target for the microphthalmia-associated transcription factor in melanocytes. Carreira S(1), Liu B, Goding CR. Author information: (1)Eukaryotic Transcription Laboratory, Marie Curie Research Institute, Oxted, Surrey RH8 OTL, United Kingdom. Commitment to the melanocyte lineage is characterized by the onset of microphthalmia-associated transcription factor (Mitf) expression. Mitf plays a fundamental role in melanocyte development, with mice lacking Mitf being entirely devoid of pigment cells. In the absence of functional Mitf protein, melanoblasts expressing Mitf mRNA disappear around 2 days after their first appearance either by apoptosis or by losing their identity and adopting an alternative cell fate. The role of Mitf must therefore be to regulate genes required for melanoblast survival, proliferation, or the maintenance of melanoblast identity. Yet to date, Mitf has been shown to regulate genes such as Tyrosinase, Tyrp-1, and Dct, which are required for pigmentation, a differentiation-specific process. Because expression of these genes cannot account for the complete absence of pigment cells in Mitf-negative mice, Mitf must regulate the expression of other as yet uncharacterized genes. Here we provide several lines of evidence to suggest that Mitf may regulate the expression of the Tbx2 transcription factor, a member of the T-box family of proteins implicated in the maintenance of cell identity. First, isolation and sequencing of the entire murine Tbx2 gene revealed that the Tbx2 promoter contains a full consensus Mitf recognition element; second, Mitf could bind the promoter in vitro and activate Tbx2 expression in vivo in an E box-dependent fashion; and third, Tbx2 is expressed in melanoma cell lines expressing Mitf, but not in a line in which Mitf expression was not detectable. Taken together, with the fact that Tbx2 is expressed in Mitf-positive melanoblasts and melanocytes, but not in Mitf-negative melanoblast precursor cells, the evidence suggests that the Tbx2 gene may represent one of the first known targets for Mitf that is not a gene involved directly in the manufacture of pigment. DOI: 10.1074/jbc.M000035200 PMID: 10770922 [Indexed for MEDLINE]