The gut microbiome: an under-recognised contributor to the COVID-19 pandemic?

Jonathan P Segal, Joyce W Y Mak, Benjamin H Mullish, James L Alexander, Siew C Ng, Julian R Marchesi, Jonathan P Segal, Joyce W Y Mak, Benjamin H Mullish, James L Alexander, Siew C Ng, Julian R Marchesi

Abstract

The novel coronavirus infection (COVID-19) caused by the SARS-CoV-2 virus has spread rapidly across the globe, culminating in major global morbidity and mortality. As such, there has been a rapid escalation in scientific and clinical activity aimed at increasing our comprehension of this virus. This volume of work has led to early insights into risk factors associated with severity of disease, and mechanisms that underpin the virulence and dynamics involved in viral transmission. These insights ultimately may help guide potential therapeutics to reduce the human, economic and social impact of this pandemic. Importantly, the gastrointestinal (GI) tract has emerged as an important organ influencing propensity to, and potentially severity of, COVID-19 infection. Furthermore, the gut microbiome has been linked to a variety of risk factors for COVID-19 infection, and manipulation of the gut microbiome is an attractive potential therapeutic target for a number of diseases. While data profiling the gut microbiome in COVID-19 infection to date are limited, they support the possibility of several routes of interaction between COVID-19, the gut microbiome, angiotensin converting enzyme 2 (ACE-2) expression in the small bowel and colon and gut inflammation. This article will explore the evidence that implicates the gut microbiome as a contributing factor to the pathogenesis, severity and disease course of COVID-19, and speculate about the gut microbiome's capability as a therapeutic avenue against COVID-19.

Lay summary: It has been noted that certain baseline gut profiles of COVID-19 patients are associated with a more severe disease course, and the gut microbiome impacts the disease course of several contributory risk factors to the severity of COVID-19. A protein called ACE-2, which is found in the small intestine among other sites, is a key receptor for COVID-19 virus entry; there is evidence that the gut microbiome influences ACE-2 receptor expression, and hence may play a role in influencing COVID-19 infectivity and disease severity. Furthermore, the gut microbiome plays a significant role in immune regulation, and hence may be pivotal in influencing the immune response to COVID-19. In terms of understanding COVID-19 treatments, the gut microbiome is known to interact with several drug classes being used to target COVID-19 and should be factored into our understanding of how patients respond to treatment. Importantly, our understanding of the role of the gut microbiome in COVID-19 infection remains in its infancy, but future research may potentially aid our mechanistic understanding of viral infection, and new ways in which we might approach treating it.

Keywords: COVID-19; SARS-CoV-2; gastrointestinal disease; microbiota.

Conflict of interest statement

Conflict of interest statement: The authors declare that there is no conflict of interest.

© The Author(s), 2020.

Figures

Figure 1.
Figure 1.
Potential mechanisms of interaction between the gut microbiome and SARS-CoV-2. SCFA, short chain fatty acid; TLR, Toll-like receptor.

References

    1. Guan W, Ni Z, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 2020; 382: 1708–1720.
    1. Zhang X, Tan Y, Ling Y, et al. Viral and host factors related to the clinical outcome of COVID-19. Nature 2020; 583: 437–440.
    1. Jin X, Lian J-S, Hu J-H, et al. Epidemiological, clinical and virological characteristics of 74 cases of coronavirus-infected disease 2019 (COVID-19) with gastrointestinal symptoms. Gut 2020; 69: 1002–1009.
    1. Lin L, Jiang X, Zhang Z, et al. Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut. Epub ahead of print 2 April 2020. DOI: 10.1136/gutjnl-2020-321013.
    1. Xiao F, Tang M, Zheng X, et al. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020; 158: 1831–1833.e3.
    1. Zheng S, Fan J, Yu F, et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January-March 2020: retrospective cohort study. BMJ 2020; 369: m1443.
    1. Effenberger M, Grabherr F, Mayr L, et al. Faecal calprotectin indicates intestinal inflammation in COVID-19. Gut. Epub ahead of print 20 April 2020. DOI: 10.1136/gutjnl-2020-321388.
    1. Lamers MM, Beumer J, van der Vaart J, et al. SARS-CoV-2 productively infects human gut enterocytes. Science. Epub ahead of print 1 May 2020. DOI: 10.1126/SCIENCE.ABC1669.
    1. Zang R, Gomez Castro MF, McCune BT, et al. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci Immunol 2020; 5: eabc3582.
    1. Gu S, Chen Y, Wu Z, et al. Alterations of the gut microbiota in patients with COVID-19 or H1N1 influenza. Clin Infect Dis. Epub ahead of print 4 June 2020. DOI: 10.1093/cid/ciaa709.
    1. Zuo T, Zhang F, Lui GCY, et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. Epub ahead of print 20 May 2020. DOI: 10.1053/J.GASTRO.2020.05.048.
    1. Geva-Zatorsky N, Sefik E, Kua L, et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 2017; 168: 928–943.e11.
    1. Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci 2008; 105: 16731–16736.
    1. Zuo T, Liu Q, Zhang F, et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut. Epub ahead of print 20 July 2020. DOI: 10.1136/gutjnl-2020-322294.
    1. Zuo T, Zhan H, Zhang F, et al. Alterations in fecal fungal microbiome of patients with COVID-19 during time of hospitalization until discharge. Gastroenterology. Epub ahead of print 26 June 2020. DOI: 10.1053/j.gastro.2020.06.048.
    1. Li M-Y, Li L, Zhang Y, et al. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect Dis Poverty 2020; 9: 45.
    1. Jordan RE, Adab P, Cheng KK. COVID-19: risk factors for severe disease and death. BMJ 2020; 368: m1198.
    1. Claesson MJ, Jeffery IB, Conde S, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature 2012; 488: 178–184.
    1. Odamaki T, Kato K, Sugahara H, et al. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol 2016; 16: 90.
    1. O’Toole PW, Jeffery IB. Gut microbiota and aging. Science 2015; 350: 1214–1215.
    1. Jackson MA, Jeffery IB, Beaumont M, et al. Signatures of early frailty in the gut microbiota. Genome Med 2016; 8: 8.
    1. Claesson MJ, Cusack S, O’Sullivan O, et al. Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci 2011; 108: 4586–4591.
    1. Kim S, Jazwinski SM. The gut microbiota and healthy aging: a mini-review. Gerontology 2018; 64: 513–520.
    1. Enaud R, Prevel R, Ciarlo E, et al. The gut-lung axis in health and respiratory diseases: a place for inter-organ and inter-kingdom crosstalks. Front Cell Infect Microbiol 2020; 10: 9.
    1. Groves HT, Cuthbertson L, James P, et al. Respiratory disease following viral lung infection alters the murine gut microbiota. Front Immunol 2018; 9: 182.
    1. Brooks AW, Priya S, Blekhman R, et al. Gut microbiota diversity across ethnicities in the United States. PLoS Biol 2018; 16: e2006842.
    1. Kumar A, Arora A, Sharma P, et al. Is diabetes mellitus associated with mortality and severity of COVID-19? A meta-analysis. Diabetes Metab Syndr 2020; 14: 535–545.
    1. Tilg H, Moschen AR. Microbiota and diabetes: an evolving relationship. Gut 2014; 63: 1513–1521.
    1. Larsen N, Vogensen FK, van den Berg FWJ, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One 2010; 5: e9085.
    1. Gurung M, Li Z, You H, et al. Role of gut microbiota in type 2 diabetes pathophysiology. EBioMedicine 2020; 51: 102590.
    1. Yadav H, Jain S, Sinha PR. Antidiabetic effect of probiotic dahi containing Lactobacillus acidophilus and Lactobacillus casei in high fructose fed rats. Nutrition 2007; 23: 62–68.
    1. Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, et al. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 2012; 28: 539–543.
    1. Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395: 497–506.
    1. Zhang J-J, Dong X, Cao Y-Y, et al. Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan, China. Allergy. Epub ahead of print 19 February 2020. DOI: 10.1111/all.14238.
    1. Lippi G, Wong J, Henry BM. Hypertension in patients with coronavirus disease 2019 (COVID-19): a pooled analysis. Polish Arch Intern Med 2020; 130: 304–309.
    1. Santisteban MM, Qi Y, Zubcevic J, et al. Hypertension-linked pathophysiological alterations in the gut. Circ Res 2017; 120: 312–323.
    1. Yan Q, Gu Y, Li X, et al. Alterations of the gut microbiome in hypertension. Front Cell Infect Microbiol 2017; 7: 381.
    1. Sun S, Lulla A, Sioda M, et al. Gut microbiota composition and blood pressure. Hypertension 2019; 73: 998–1006.
    1. Seppo L, Jauhiainen T, Poussa T, et al. A fermented milk high in bioactive peptides has a blood pressure-lowering effect in hypertensive subjects. Am J Clin Nutr 2003; 77: 326–330.
    1. Cole-Jeffrey CT, Liu M, Katovich MJ, et al. ACE2 and microbiota: emerging targets for cardiopulmonary disease therapy. J Cardiovasc Pharmacol 2015; 66: 540–550.
    1. Sriram K, Insel PA. Risks of ACE inhibitor and ARB usage in COVID-19: evaluating the evidence. Clin Pharmacol Ther. Epub ahead of print 22 April 2020. DOI: 10.1002/cpt.1863.
    1. de Abajo FJ, Rodríguez-Martín S, Lerma V, et al. Use of renin-angiotensin-aldosterone system inhibitors and risk of COVID-19 requiring admission to hospital: a case-population study. Lancet 2020; 395: 1705–1714.
    1. Kirby T. Evidence mounts on the disproportionate effect of COVID-19 on ethnic minorities. Lancet Respir Med 2020; 8: 547–548.
    1. Deschasaux M, Bouter KE, Prodan A, et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat Med 2018; 24: 1526–1531.
    1. He Y, Wu W, Zheng H-M, et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat Med 2018; 24: 1532–1535.
    1. Dhakan DB, Maji A, Sharma AK, et al. The unique composition of Indian gut microbiome, gene catalogue, and associated fecal metabolome deciphered using multi-omics approaches. Gigascience 2019; 8: giz004.
    1. Hashimoto T, Perlot T, Rehman A, et al. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature 2012; 487: 477–481.
    1. Wang J, Zhao S, Liu M, et al. ACE2 expression by colonic epithelial cells is associated with viral infection, immunity and energy metabolism. medRxiv 2020.02.05.20020545. DOI: 10.1101/2020.02.05.20020545.
    1. Yang T, Chakraborty S, Saha P, et al. Gnotobiotic rats reveal that gut microbiota regulates colonic mRNA of Ace2, the receptor for SARS-CoV-2 infectivity. Hypertension. Epub ahead of print 19 May 2020. DOI: 10.1161/HYPERTENSIONAHA.120.15360.
    1. Vuille-dit-Bille RN, Liechty KW, Verrey F, et al. SARS-CoV-2 receptor ACE2 gene expression in small intestine correlates with age. Amino Acids 2020; 52: 1063–1065.
    1. Zhang H, Kang Z, Gong H, et al. Digestive system is a potential route of COVID-19: an analysis of single-cell coexpression pattern of key proteins in viral entry process. Gut 2020; 69: 1010–1018.
    1. Hamming I, Timens W, Bulthuis MLC, et al. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 2004; 203: 631–637.
    1. Garg M, Burrell LM, Velkoska E, et al. Upregulation of circulating components of the alternative renin-angiotensin system in inflammatory bowel disease: a pilot study. J Renin Angiotensin Aldosterone Syst 2015; 16: 559–569.
    1. Zuo T, Ng SC. The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease. Front Microbiol 2018; 9: 2247.
    1. Shen Z, Xiao Y, Kang L, et al. Genomic diversity of severe acute respiratory syndrome-coronavirus 2 in patients with coronavirus disease 2019. Clin Infect Dis 2020; 71: 713–720.
    1. Fan J, Li X, Gao Y, et al. The lung tissue microbiota features of 20 deceased patients with COVID-19. J Infect 2020; 81: e64–e67.
    1. Diao B, Wang C, Tan Y, et al. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol 2020; 11: 827.
    1. Tanoue T, Morita S, Plichta DR, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 2019; 565: 600–605.
    1. Yu AI, Zhao L, Eaton KA, et al. Gut microbiota modulate CD8 T cell responses to influence colitis-associated tumorigenesis. Cell Rep 2020; 31: 107471.
    1. Bachem A, Makhlouf C, Binger KJ, et al. Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8+ T Cells. Immunity 2019; 51: 285–297.e5.
    1. Luu M, Weigand K, Wedi F, et al. Regulation of the effector function of CD8+ T cells by gut microbiota-derived metabolite butyrate. Sci Rep 2018; 8: 14430.
    1. Virgin HW. The virome in mammalian physiology and disease. Cell 2014; 157: 142–150.
    1. Weaver LK, Minichino D, Biswas C, et al. Microbiota-dependent signals are required to sustain TLR-mediated immune responses. JCI Insight 2019; 4: e124370.
    1. Mukhopadhya I, Segal JP, Carding SR, et al. The gut virome: the ‘missing link’ between gut bacteria and host immunity? Therap Adv Gastroenterol 2019; 12: 175628481983662.
    1. Monedero V, Buesa J, Rodríguez-Díaz J. The interactions between host glycobiology, bacterial microbiota, and viruses in the gut. Viruses 2018; 10: 96.
    1. Allen IC, Scull MA, Moore CB, et al. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity 2009; 30: 556–565.
    1. Ichinohe T, Lee HK, Ogura Y, et al. Inflammasome recognition of influenza virus is essential for adaptive immune responses. J Exp Med 2009; 206: 79–87.
    1. Ilie PC, Stefanescu S, Smith L. The role of vitamin D in the prevention of coronavirus disease 2019 infection and mortality. Aging Clin Exp Res. Epub ahead of print 6 May 2020. DOI: 10.1007/S40520-020-01570-8.
    1. Rhodes JM, Subramanian S, Laird E, et al. Editorial: low population mortality from COVID-19 in countries South of latitude 35 degrees North supports vitamin D as a factor determining severity. Aliment Pharmacol Ther. Epub ahead of print 28 April 2020. DOI: 10.1111/apt.15777.
    1. Baktash V, Hosack T, Patel N, et al. Vitamin D status and outcomes for hospitalised older patients with COVID-19. Postgrad Med J. Epub ahead of print 27 August 2020. DOI: 10.1136/postgradmedj-2020-138712.
    1. Entrenas Castillo M, Entrenas Costa LM, Vaquero Barrios JM, et al. Effect of calcifediol treatment and best available therapy versus best available therapy on intensive care unit admission and mortality among patients hospitalized for COVID-19: a pilot randomized clinical study. J Steroid Biochem Mol Biol 2020; 203: 105751.
    1. Garg M, Al-Ani A, Mitchell H, et al. Editorial: low population mortality from COVID-19 in countries South of latitude 35 degrees North-supports vitamin D as a factor determining severity. Authors’ reply. Aliment Pharmacol Ther. Epub ahead of print 12 May 2020. DOI: 10.1111/apt.15796.
    1. Hart PH, Gorman S, Finlay-Jones JJ. Modulation of the immune system by UV radiation: more than just the effects of vitamin D? Nat Rev Immunol 2011; 11: 584–596.
    1. Yamamoto E, Jørgensen TN. Immunological effects of vitamin D and their relations to autoimmunity. J Autoimmun 2019; 100: 7–16.
    1. Luthold RV, Fernandes GR, Franco-de-Moraes AC, et al. Gut microbiota interactions with the immunomodulatory role of vitamin D in normal individuals. Metabolism 2017; 69: 76–86.
    1. Ooi JH, Li Y, Rogers CJ, et al. Vitamin D regulates the gut microbiome and protects mice from dextran sodium sulfate–induced colitis. J Nutr 2013; 143: 1679–1686.
    1. Makishima M, Lu TT, Xie W, et al. Vitamin D receptor as an intestinal bile acid sensor. Science 2002; 296: 1313–1316.
    1. Dofferhoff ASM, Piscaer I, Schurgers LJ, et al. Reduced vitamin K status as a potentially modifiable prognostic risk factor in COVID-19. Clin Infect Dis. Epub ahead of print 25 April 2020. DOI: 10.20944/PREPRINTS202004.0457.V1.
    1. Beulens JWJ, Booth SL, van den Heuvel EGHM, et al. The role of menaquinones (vitamin K2) in human health. Br J Nutr 2013; 110: 1357–1368.
    1. Maggini S, Pierre A, Calder PC. Immune function and micronutrient requirements change over the life course. Nutrients 2018; 10: 1531.
    1. Butler MJ, Barrientos RM. The impact of nutrition on COVID-19 susceptibility and long-term consequences. Brain Behav Immun. Epub ahead of print 18 April 2020. DOI: 10.1016/J.BBI.2020.04.040.
    1. World Health Organization. Food and nutrition tips during self-quarantine, (accessed 19 May 2020).
    1. Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol 2019; 16: 35–56.
    1. National Health Committee of the People’s Republic of China, National Administration of Traditional Chinese Medicine and National Health Committee of the People’s Republic of China. National administration of traditional Chinese medicine diagnostic and therapeutic guidance for 2019 Novel coronavirus disease. (version 5).
    1. Morrow LE, Kollef MH, Casale TB. Probiotic prophylaxis of ventilator-associated pneumonia: a blinded, randomized, controlled trial. Am J Respir Crit Care Med 2010; 182: 1058–1064.
    1. Zeng J, Wang C-T, Zhang F-S, et al. Effect of probiotics on the incidence of ventilator-associated pneumonia in critically ill patients: a randomized controlled multicenter trial. Intensive Care Med 2016; 42: 1018–1028.
    1. Mak JWY, Chan FKL, Ng SC. Probiotics and COVID-19: one size does not fit all. Lancet Gastroenterol Hepatol. Epub ahead of print 25 April 2020. DOI: 10.1016/S2468-1253(20)30122-9.
    1. Weblet Importer. Global coronavirus COVID-19 clinical trial tracker, (accessed 2 June 2020).
    1. Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, et al. Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature 2019; 570: 462–467.
    1. Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, et al. Separating host and microbiome contributions to drug pharmacokinetics and toxicity. Science 2019; 363: eaat9931.
    1. Forslund K, Hildebrand F, Nielsen T, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015; 528: 262–266.
    1. Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018; 359: 104–108.
    1. Engberg J, Aarestrup FM, Taylor DE, et al. Quinolone and macrolide resistance in campylobacter jejuni and C. Coli: resistance mechanisms and trends in human isolates. Emerg Infect Dis 2001; 7: 24–34.
    1. Wei S, Mortensen MS, Stokholm J, et al. Short- and long-term impacts of azithromycin treatment on the gut microbiota in children: a double-blind, randomized, placebo-controlled trial. EBioMedicine 2018; 38: 265–272.
    1. Doan T, Hinterwirth A, Worden L, et al. Gut microbiome alteration in MORDOR I: a community-randomized trial of mass azithromycin distribution. Nat Med 2019; 25: 1370–1376.
    1. Abeles SR, Jones MB, Santiago-Rodriguez TM, et al. Microbial diversity in individuals and their household contacts following typical antibiotic courses. Microbiome 2016; 4: 39.
    1. Strathdee SA, Davies SC, Marcelin JR. Confronting antimicrobial resistance beyond the COVID-19 pandemic and the 2020 US election. Lancet 2020; 396: 1050–1053.
    1. Bradley KC, Finsterbusch K, Schnepf D, et al. Microbiota-driven tonic interferon signals in lung stromal cells protect from influenza virus infection. Cell Rep 2019; 28: 245–256.e4.
    1. Baud D, Dimopoulou Agri V, Gibson GR, et al. Using probiotics to flatten the curve of coronavirus disease COVID-2019 pandemic. Front public Health 2020; 8: 186.
    1. McIlroy JR, Mullish BH, Goldenberg SD, et al. Intestinal microbiome transfer, a novel therapeutic strategy for COVID-19 induced hyperinflammation?: in reply to, ‘COVID-19: immunology and treatment options’, Felsenstein, Herbert McNamara et al. 2020’. Clin Immunol 2020; 218: 108542.
    1. Zhang C, Wu Z, Li J-W, et al. Cytokine release syndrome in severe COVID-19: interleukin-6 receptor antagonist tocilizumab may be the key to reduce mortality. Int J Antimicrob Agents 2020; 55: 105954.
    1. Vikse J, Henry BM. Tocilizumab in COVID-19: beware the risk of intestinal perforation. Int J Antimicrob Agents 2020; 56: 106009.
    1. Strangfeld A, Richter A, Siegmund B, et al. Risk for lower intestinal perforations in patients with rheumatoid arthritis treated with tocilizumab in comparison to treatment with other biologic or conventional synthetic DMARDs. Ann Rheum Dis 2017; 76: 504–510.
    1. Barbara G, Scaioli E, Barbaro MR, et al. Gut microbiota, metabolome and immune signatures in patients with uncomplicated diverticular disease. Gut 2017; 66: 1252–1261.
    1. Kuhn KA, Schulz HM, Regner EH, et al. Bacteroidales recruit IL-6-producing intraepithelial lymphocytes in the colon to promote barrier integrity. Mucosal Immunol 2018; 11: 357–368.
    1. Tsang JS, Dobaño C, VanDamme P, et al. Improving vaccine-induced immunity: can baseline predict outcome? Trends Immunol 2020; 41: 457–465.
    1. Hagan T, Cortese M, Rouphael N, et al. Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans. Cell 2019; 178: 1313–1328.e13.
    1. Wu Y, Guo C, Tang L, et al. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol Hepatol 2020; 5: 434–435.
    1. Iacucci M, Cannatelli R, Labarile N, et al. Endoscopy in inflammatory bowel diseases during the COVID-19 pandemic and post-pandemic period. Lancet Gastroenterol Hepatol 2020; 5: 598–606.
    1. Al-Jabir A, Kerwan A, Nicola M, et al. Impact of the coronavirus (COVID-19) pandemic on surgical practice - part 1. Int J Surg. Epub ahead of print 12 May 2020. DOI: 10.1016/J.IJSU.2020.05.022.
    1. Haleem A, Javaid M, Vaishya R, et al. Areas of academic research with the impact of COVID-19. Am J Emerg Med. Epub ahead of print 15 April 2020. DOI: 10.1016/J.AJEM.2020.04.022.
    1. Gallo G, Trompetto M. The effects of COVID-19 on academic activities and surgical education in Italy. J Investig Surg 2020; 33: 687–689.
    1. Rawson TM, Moore LSP, Castro-Sanchez E, et al. COVID-19 and the potential long-term impact on antimicrobial resistance. J Antimicrob Chemother. Epub ahead of print 20 May 2020. DOI: 10.1093/jac/dkaa194.

Source: PubMed

3
Předplatit