Generic placeholder image

Infectious Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5265
ISSN (Online): 2212-3989

Review Article

An Updated Review on Complicated Mechanisms of COVID-19 Pathogenesis and Therapy: Direct Viral Damage, Renin-angiotensin System Dysregulation, Immune System Derangements, and Endothelial Dysfunction

Author(s): Shahab Falahi, Maryam Maleki and Azra Kenarkoohi*

Volume 22, Issue 7, 2022

Published on: 04 August, 2022

Article ID: e210322202494 Pages: 11

DOI: 10.2174/1871526522666220321153712

Price: $65

Abstract

SARS-CoV-2 was reported as the cause of coronavirus disease 2019 (COVID-19) in late December 2019. According to sequencing and phylogenetic studies, the new virus belongs to Coronaviridae family and Betacoronavirus genus. Genomic sequence analysis has shown SARS-CoV-2 to be similar to SARS. SARS-CoV-2 is more infectious, and the high level of COVID-19 community transmission has led to a growing pandemic. Although infections in most patients with COVID-19 are moderate or mild, 20% of the patients develop a severe or critical form of the disease. COVID-19 may affect a wide range of organs and tissues, including the respiratory system, digestive system, nervous system, and skin. Patients with COVID-19 have been confirmed to have renal, cardiovascular, gastrointestinal, and nervous system problems in addition to pulmonary involvement. The pathogenesis of SARS-CoV-2 is being investigated, but it is possible that the organ damage might in part be caused by direct viral damage (detection of inclusion bodies in tissues, such as the kidneys), dysregulation of the immune system, renin-angiotensin system, bradykinin pathway, and coagulation, as well as host genetic factors and their polymorphisms, which may affect the disease severity. In this review, an update on the possible pathogenesis pathways of COVID-19 has been provided. It is hoped that the best care strategy will be developed for patients with COVID-19 by identifying its pathogenesis pathways.

Keywords: SARS-CoV-2, COVID-19, pathogenesis, Renin-angiotensin system, thromboinflammation, immunopathogenesis.

Graphical Abstract
[1]
Gupta A, Madhavan MV, Sehgal K, et al. Extrapulmonary manifestations of COVID-19. Nat Med 2020; 26(7): 1017-32.
[http://dx.doi.org/10.1038/s41591-020-0968-3] [PMID: 32651579]
[2]
Dockery DM, Rowe SG, Murphy MA, Krzystolik MG. The Ocular manifestations and transmission of COVID-19: Recommendations for prevention. J Emerg Med 2020; 59(1): 137-40.
[http://dx.doi.org/10.1016/j.jemermed.2020.04.060] [PMID: 32456959]
[3]
Abdoli A, Falahi S, Kenarkoohi A, Shams M, Mir H, Jahromi MAM. The COVID-19 pandemic, psychological stress during pregnancy, and risk of neurodevelopmental disorders in offspring: A neglected consequence. J Psychosom Obstet Gynaecol 2020; 41(3): 247-8.
[http://dx.doi.org/10.1080/0167482X.2020.1761321] [PMID: 32380881]
[4]
Mohamed AA, Mohamed N, Mohamoud S, et al. SARS-CoV-2: The path of prevention and control. Infectious Disorders-Drug Targets 2021; 21(3): 358-62.
[PMID: 32433010]
[5]
Mohammadyari E, Kaffashian MR, Ahmadi I, et al. Clinical features of novel coronavirus 2019-infected cases with pre-existing cardiovas-cular disease, disaggregated by gender. Pak Heart J 2021; 54(2): 180-5.
[http://dx.doi.org/10.47144/phj.v54i2.2096]
[6]
Sadeghifar J, Jalilian H, Momeni K, et al. Outcome evaluation of COVID-19 infected patients by disease symptoms: A cross-sectional study in Ilam province, Iran. BMC Infect Dis 2021; 21(1): 903.
[http://dx.doi.org/10.1186/s12879-021-06613-7] [PMID: 34479500]
[7]
Goyal P, Choi JJ, Pinheiro LC, et al. Clinical characteristics of COVID-19 in New York city. N Engl J Med 2020; 382(24): 2372-4.
[http://dx.doi.org/10.1056/NEJMc2010419] [PMID: 32302078]
[8]
Zhu J, Ji P, Pang J, et al. Clinical characteristics of 3062 COVID-19 patients: A meta-analysis. J Med Virol 2020; 92(10): 1902-14.
[http://dx.doi.org/10.1002/jmv.25884] [PMID: 32293716]
[9]
Zheng Z, Peng F, Xu B, et al. Risk factors of critical & mortal COVID-19 cases: A systematic literature review and meta-analysis. J Infect 2020; 81(2): e16-25.
[http://dx.doi.org/10.1016/j.jinf.2020.04.021] [PMID: 32335169]
[10]
Zhou Y, Chi J, Lv W, Wang Y. Obesity and diabetes as high-risk factors for severe coronavirus disease 2019 (COVID-19). Diabetes Metab Res Rev 2021; 37(2): e3377.
[http://dx.doi.org/10.1002/dmrr.3377] [PMID: 32588943]
[11]
Benskin LL. A basic review of the preliminary evidence that COVID-19 risk and severity is increased in vitamin D deficiency. Front Public Health 2020; 8: 513.
[http://dx.doi.org/10.3389/fpubh.2020.00513] [PMID: 33014983]
[12]
Falahi S, Kenarkoohi A. Sex and gender differences in the outcome of patients with COVID-19. J Med Virol 2021; 93(1): 151-2.
[http://dx.doi.org/10.1002/jmv.26243] [PMID: 32603509]
[13]
Wei Y-Y, Wang R-R, Zhang D-W, et al. Risk factors for severe COVID-19: Evidence from 167 hospitalized patients in Anhui, China. J Infect 2020; 81(1): e89-92.
[http://dx.doi.org/10.1016/j.jinf.2020.04.010] [PMID: 32305487]
[14]
Lippi G, Plebani M, Henry BM. Thrombocytopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: A meta-analysis. Clin Chim Acta 2020; 506: 145-8.
[PMID: 32178975]
[15]
Liao D, Zhou F, Luo L, et al. Haematological characteristics and risk factors in the classification and prognosis evaluation of COVID-19: A retrospective cohort study. Lancet Haematol 2020; 7(9): e671-8.
[http://dx.doi.org/10.1016/S2352-3026(20)30217-9] [PMID: 32659214]
[16]
Kenarkoohi A, Noorimotlagh Z, Falahi S, et al. Hospital indoor air quality monitoring for the detection of SARS-CoV-2 (COVID-19) virus. Sci Total Environ 2020; 748: 141324.
[http://dx.doi.org/10.1016/j.scitotenv.2020.141324] [PMID: 32805566]
[17]
Falahi S, Kenarkoohi A. COVID-19 reinfection: prolonged shedding or true reinfection? New Microbes New Infect 2020; 38: 100812.
[http://dx.doi.org/10.1016/j.nmni.2020.100812] [PMID: 33200033]
[18]
Falahi S, Kenarkoohi A. Transmission routes for SARS-CoV-2 infection: Review of evidence. New Microbes New Infect 2020; 38: 100778.
[http://dx.doi.org/10.1016/j.nmni.2020.100778] [PMID: 33042554]
[19]
Kenarkoohi A, Maleki M, Safari T, Kafashian M, Saljoughi F, Sohrabipour S. Angiotensin-converting enzyme 2 roles in the pathogenesis of COVID-19. Curr Hypertens Rev 2021; 17(3): 207-16.
[PMID: 32778033]
[20]
Falahi S, Abdoli A, Kenarkoohi A. Claims and reasons about mild COVID-19 in children. New Microbes New Infect 2021; 41: 100864.
[http://dx.doi.org/10.1016/j.nmni.2021.100864] [PMID: 33747533]
[21]
Kenar Koohi A, Ravanshad M, Rasouli M, Falahi S, Baghban A. Phylogenetic analysis of torque teno virus in hepatitis C virus infected patients in Shiraz. Hepat Mon 2012; 12(7): 437-41.
[http://dx.doi.org/10.5812/hepatmon.6133] [PMID: 23008723]
[22]
Mirrnejad R, Fallahi S, Kiani J, et al. Epidemic assessment of bacterial agents in osteomyelitis and their antibiotic resistance pattern deter-mination. J Biol Sci (Faisalabad, Pak) 2008; 8(2): 478-81.
[http://dx.doi.org/10.3923/jbs.2008.478.481]
[23]
Kenarkoohi A, Bamdad T, Soleimani M, Soleimanjahi H, Fallah A, Falahi S. HSV-TK Expressing mesenchymal stem cells exert inhibitory effect on cervical cancer model. Int J Mol Cell Med 2020; 9(2): 146-54.
[PMID: 32934952]
[24]
Abdoli A, Taghipour A, Pirestani M, et al. Infections, inflammation, and risk of neuropsychiatric disorders: The neglected role of “co-infection”. Heliyon 2020; 6(12): e05645.
[http://dx.doi.org/10.1016/j.heliyon.2020.e05645] [PMID: 33319101]
[25]
Ravanshad M, Sabahi F, Falahi S. Prediction of hepatitis B virus lamivudine resistance based on YMDD sequence data using an artificial neural network model. Hepat Mon 2011; 11(2): 108-13.
[26]
Mason RJ. Pathogenesis of COVID-19 from a cell biology perspective. Eur Respir J 2020; 55(4): 2000607.
[http://dx.doi.org/10.1183/13993003.00607-2020] [PMID: 32269085]
[27]
Kim JM, Chung YS, Jo HJ, et al. Identification of coronavirus isolated from a patient in Korea with COVID-19. Osong Public Health Res Perspect 2020; 11(1): 3-7.
[http://dx.doi.org/10.24171/j.phrp.2020.11.1.02] [PMID: 32149036]
[28]
Maggi E, Canonica GW, Moretta L. COVID-19: Unanswered questions on immune response and pathogenesis. J Allergy Clin Immunol 2020; 146(1): 18-22.
[http://dx.doi.org/10.1016/j.jaci.2020.05.001] [PMID: 32389590]
[29]
Pan X-W, Xu D, Zhang H, Zhou W, Wang L-H, Cui X-G. Identification of a potential mechanism of acute kidney injury during the COVID-19 outbreak: A study based on single-cell transcriptome analysis. Intensive Care Med 2020; 46(6): 1114-6.
[http://dx.doi.org/10.1007/s00134-020-06026-1] [PMID: 32236644]
[30]
Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020; 395(10234): 1417-8.
[http://dx.doi.org/10.1016/S0140-6736(20)30937-5] [PMID: 32325026]
[31]
Capuano I, Buonanno P, Riccio E, Pisani A. Acute kidney injury in COVID-19 pandemic. Nephron 2020; 144(7): 345-6.
[http://dx.doi.org/10.1159/000508381] [PMID: 32428921]
[32]
Kenarkoohi A, Maleki M, Ghiasi B, et al. Prevalence and clinical presentation of COVID-19 infection in hemodialysis patients. J Nephropathol 2022; 11(1): 1-16.
[33]
Simsek-Yavuz S, Komsuoglu Celikyurt FI. Antiviral treatment of COVID-19: An update. Turk J Med Sci 2020; 50(SI-1): 611-9.
[PMID: 32293834]
[34]
Stojkovic-Filipovic J, Bosic M. Treatment of COVID 19-repurposing drugs commonly used in dermatology. Dermatol Ther 2020; 33(5): e13829.
[http://dx.doi.org/10.1111/dth.13829] [PMID: 32542964]
[35]
Cao W, Li T. COVID-19: towards understanding of pathogenesis. Cell Res 2020; 30(5): 367-9.
[http://dx.doi.org/10.1038/s41422-020-0327-4] [PMID: 32346073]
[36]
Zhu H, Rhee J-W, Cheng P, et al. Cardiovascular complications in patients with COVID-19: Consequences of viral toxicities and host im-mune response. Curr Cardiol Rep 2020; 22(5): 32.
[http://dx.doi.org/10.1007/s11886-020-01292-3] [PMID: 32318865]
[37]
Bertoli F, Veritti D, Danese C, et al. Ocular findings in COVID-19 patients: A review of direct manifestations and indirect effects on the eye. J Ophthalmol 2020; 2020: 4827304.
[http://dx.doi.org/10.1155/2020/4827304] [PMID: 32963819]
[38]
Rosa SGV, Santos WC. Clinical trials on drug repositioning for COVID-19 treatment. Rev Panam Salud Publica 2020; 44: e40.
[http://dx.doi.org/10.26633/RPSP.2020.40] [PMID: 32256547]
[39]
Tahir Ul Qamar M, Alqahtani SM, Alamri MA, Chen LL. Structural basis of SARS-CoV-2 3CLpro and anti-COVID-19 drug discovery from medicinal plants. J Pharm Anal 2020; 10(4): 313-9.
[http://dx.doi.org/10.1016/j.jpha.2020.03.009] [PMID: 32296570]
[40]
Singh AK, Singh A, Singh R, Misra A. Molnupiravir in COVID-19: A systematic review of literature. Diabetes Metab Syndr 2021; 15(6): 102329.
[http://dx.doi.org/10.1016/j.dsx.2021.102329] [PMID: 34742052]
[41]
Imran M, Kumar Arora M, Asdaq SMB, et al. Discovery, development, and patent trends on molnupiravir: A prospective oral treatment for COVID-19. Molecules 2021; 26(19): 5795.
[http://dx.doi.org/10.3390/molecules26195795] [PMID: 34641339]
[42]
Abd-Elsalam S, Ahmed OA, Mansour NO, et al. Remdesivir efficacy in COVID-19 treatment: A randomized controlled trial. Am J Trop Med Hyg 2021; 106(3): 886.
[http://dx.doi.org/10.4269/ajtmh.21-0606] [PMID: 34649223]
[43]
Dabbous HM, Abd-Elsalam S, El-Sayed MH, et al. Efficacy of favipiravir in COVID-19 treatment: A multi-center randomized study. Arch Virol 2021; 166(3): 949-54.
[http://dx.doi.org/10.1007/s00705-021-04956-9] [PMID: 33492523]
[44]
El-Bendary M, Abd-Elsalam S, Elbaz T, et al. Efficacy of combined sofosbuvir and daclatasvir in the treatment of COVID-19 patients with pneumonia: A multicenter Egyptian study. Expert Rev Anti Infect Ther 2021; 1-5.
[http://dx.doi.org/10.1080/14787210.2021.1950532] [PMID: 34225541]
[45]
Abd-Elsalam S, Soliman S, Esmail ES, et al. Do zinc supplements enhance the clinical efficacy of hydroxychloroquine?: A randomized, multicenter trial. Biol Trace Elem Res 2021; 199(10): 3642-6.
[PMID: 33247380]
[46]
Abd-Elsalam S, Noor RA, Badawi R, et al. Clinical study evaluating the efficacy of ivermectin in COVID-19 treatment: A randomized con-trolled study. J Med Virol 2021; 93(10): 5833-8.
[http://dx.doi.org/10.1002/jmv.27122] [PMID: 34076901]
[47]
Edmonston DL, South AM, Sparks MA, Cohen JB. Coronavirus disease 2019 and Hypertension: The role of angiotensin-converting en-zyme 2 and the renin-angiotensin system. Adv Chronic Kidney Dis 2020; 27(5): 404-11.
[http://dx.doi.org/10.1053/j.ackd.2020.07.002] [PMID: 33308506]
[48]
Rysz S, Al-Saadi J, Sjöström A, et al. COVID-19 pathophysiology may be driven by an imbalance in the renin-angiotensin-aldosterone system. Nat Commun 2021; 12(1): 2417.
[http://dx.doi.org/10.1038/s41467-021-22713-z] [PMID: 33893295]
[49]
Wu J, Deng W, Li S, Yang X. Advances in research on ACE2 as a receptor for 2019-nCoV. Cellular and molecular life sciences. Cell Mol Life Sci 2021; 78(2): 531-44.
[PMID: 32780149]
[50]
Meng W, Zhao W, Zhao T, et al. Autocrine and paracrine function of Angiotensin 1-7 in tissue repair during hypertension. Am J Hypertens 2014; 27(6): 775-82.
[http://dx.doi.org/10.1093/ajh/hpt270] [PMID: 24429674]
[51]
Ziegler CGK, Allon SJ, Nyquist SK, et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 2020; 181(5): 1016-1035.e19.
[http://dx.doi.org/10.1016/j.cell.2020.04.035] [PMID: 32413319]
[52]
Gao YM, Xu G, Wang B, Liu BC. Cytokine storm syndrome in coronavirus disease 2019: A narrative review. J Intern Med 2020; 288(6): 1-15.
[PMID: 32696489]
[53]
Namsolleck P, Recarti C, Foulquier S, Steckelings UM, Unger T. AT(2) receptor and tissue injury: therapeutic implications. Curr Hypertens Rep 2014; 16(2): 416.
[http://dx.doi.org/10.1007/s11906-013-0416-6] [PMID: 24414230]
[54]
Aronson JK, Ferner RE. Drugs and the renin-angiotensin system in covid-19. BMJ 2020; 369: m1313.
[http://dx.doi.org/10.1136/bmj.m1313] [PMID: 32241880]
[55]
Mehta N, Kalra A, Nowacki AS, et al. Association of use of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers with testing positive for coronavirus disease 2019 (COVID-19). JAMA Cardiol 2020; 5(9): 1020-6.
[http://dx.doi.org/10.1001/jamacardio.2020.1855] [PMID: 32936273]
[56]
Mancia G, Rea F, Ludergnani M, Apolone G, Corrao G. Renin–angiotensin–aldosterone system blockers and the risk of COVID-19. N Engl J Med 2020; 382(25): 2431-40.
[http://dx.doi.org/10.1056/NEJMoa2006923] [PMID: 32356627]
[57]
Zhang P, Zhu L, Cai J, et al. Association of inpatient use of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers with mortality among patients with hypertension hospitalized with COVID-19. Circ Res 2020; 126(12): 1671-81.
[http://dx.doi.org/10.1161/CIRCRESAHA.120.317134] [PMID: 32302265]
[58]
Kuster GM, Pfister O, Burkard T, et al. SARS-CoV2: should inhibitors of the renin-angiotensin system be withdrawn in patients with COVID-19? Eur Heart J 2020; 41(19): 1801-3.
[http://dx.doi.org/10.1093/eurheartj/ehaa235] [PMID: 32196087]
[59]
Meng J, Xiao G, Zhang J, et al. Renin-angiotensin system inhibitors improve the clinical outcomes of COVID-19 patients with hyperten-sion. Emerg Microbes Infect 2020; 9(1): 757-60.
[http://dx.doi.org/10.1080/22221751.2020.1746200] [PMID: 32228222]
[60]
Garvin MR, Alvarez C, Miller JI, et al. A mechanistic model and therapeutic interventions for COVID-19 involving a RAS-mediated brady-kinin storm. eLife 2020; 9: e59177.
[http://dx.doi.org/10.7554/eLife.59177] [PMID: 32633718]
[61]
Roche JA, Roche R. A hypothesized role for dysregulated bradykinin signaling in COVID-19 respiratory complications. FASEB J 2020; 34(6): 7265-9.
[http://dx.doi.org/10.1096/fj.202000967] [PMID: 32359101]
[62]
de Maat S, de Mast Q, Danser AHJ, van de Veerdonk FL, Maas C. Impaired breakdown of bradykinin and its metabolites as a possible cause for pulmonary edema in COVID-19 infection. Semin Thromb Hemost 2020; 46(7): 835-7.
[http://dx.doi.org/10.1055/s-0040-1712960] [PMID: 32526773]
[63]
Meini S, Zanichelli A, Sbrojavacca R, et al. Understanding the pathophysiology of COVID-19: Could the contact system be the key? Front Immunol 2020; 11: 2014.
[PMID: 32849666]
[64]
Tolouian R, Zununi Vahed S, Ghiyasvand S, Tolouian A, Ardalan M. COVID-19 interactions with angiotensin-converting enzyme 2 (ACE2) and the kinin system; looking at a potential treatment. J Renal Inj Prev 2020; 9(2): e19.
[http://dx.doi.org/10.34172/jrip.2020.19]
[65]
Ghahestani SM, Mahmoudi J, Hajebrahimi S, et al. Bradykinin as a probable aspect in SARS-Cov-2 Scenarios: Is bradykinin sneaking out of our sight? Iran J Allergy Asthma Immunol 2020; 19(S1): 13-7.
[http://dx.doi.org/10.18502/ijaai.v19i(s1.r1).2850] [PMID: 32534506]
[66]
Farmer SG, Wilkins DE, Meeker SA, Seeds EA, Page CP. Effects of bradykinin receptor antagonists on antigen-induced respiratory dis-tress, airway hyperresponsiveness and eosinophilia in guinea-pigs. Br J Pharmacol 1992; 107(3): 653-9.
[http://dx.doi.org/10.1111/j.1476-5381.1992.tb14502.x] [PMID: 1335332]
[67]
Alkotaji M, Al-Zidan RN. Indomethacin: Can it counteract bradykinin effects in COVID-19 patients? Curr Pharmacol Rep 2021; 7(3): 1-5.
[http://dx.doi.org/10.1007/s40495-021-00257-6] [PMID: 33907665]
[68]
Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19 cytokine storm; what we know so far. Front Immunol 2020; 11: 1446.
[http://dx.doi.org/10.3389/fimmu.2020.01446] [PMID: 32612617]
[69]
Mangalmurti N, Hunter CA. Cytokine storms: Understanding COVID-19. Immunity 2020; 53(1): 19-25.
[http://dx.doi.org/10.1016/j.immuni.2020.06.017] [PMID: 32610079]
[70]
Rodríguez Y, Novelli L, Rojas M, et al. Autoinflammatory and autoimmune conditions at the crossroad of COVID-19. J Autoimmun 2020; 114: 102506.
[http://dx.doi.org/10.1016/j.jaut.2020.102506] [PMID: 32563547]
[71]
Sadeghzadeh-Bazargan A, Behrangi E, Goodarzi A. Cytokine storm and probable role of immunoregulatory drugs in COVID-19: A comprehensive review. Iranian J Dermatol 2020; 23(Suppl. 1 (COVID-19)): 13-8.
[http://dx.doi.org/10.22034/IJD.2020.114848]
[72]
Brunetti L, Diawara O, Tsai A, et al. Colchicine to weather the cytokine storm in hospitalized patients with COVID-19. J Clin Med 2020; 9(9): 2961.
[http://dx.doi.org/10.3390/jcm9092961] [PMID: 32937800]
[73]
Zhao M. Cytokine storm and immunomodulatory therapy in COVID-19: Role of chloroquine and anti-IL-6 monoclonal antibodies. Int J Antimicrob Agents 2020; 55(6): 105982.
[http://dx.doi.org/10.1016/j.ijantimicag.2020.105982] [PMID: 32305588]
[74]
Halacli B, Topeli A. Treatment of the cytokine storm in COVID-19. J Crit Intensive Care 2020; 11 (Suppl. 1): 36-40.
[http://dx.doi.org/10.37678/dcybd.2020.2434]
[75]
Huang Q, Wu X, Zheng X, Luo S, Xu S, Weng J. Targeting inflammation and cytokine storm in COVID-19. Pharmacol Res 2020; 159: 105051.
[http://dx.doi.org/10.1016/j.phrs.2020.105051] [PMID: 32603772]
[76]
Tang Y, Liu J, Zhang D, Xu Z, Ji J, Wen C. Cytokine storm in COVID-19: The current evidence and treatment strategies. Front Immunol 1708; 2020: 11.
[PMID: 32754163]
[77]
Omarjee L, Janin A, Perrot F, Laviolle B, Meilhac O, Mahe G. Targeting T-cell senescence and cytokine storm with rapamycin to prevent severe progression in COVID-19. Clin Immunol 2020; 216: 108464.
[http://dx.doi.org/10.1016/j.clim.2020.108464] [PMID: 32405269]
[78]
Golchin A, Seyedjafari E, Ardeshirylajimi A. Mesenchymal stem cell therapy for COVID-19: Present or future. Stem Cell Rev Rep 2020; 16(3): 427-33.
[http://dx.doi.org/10.1007/s12015-020-09973-w] [PMID: 32281052]
[79]
Sadeghi S, Soudi S, Shafiee A, Hashemi SM. Mesenchymal stem cell therapies for COVID-19: Current status and mechanism of action. Life Sci 2020; 262: 118493.
[http://dx.doi.org/10.1016/j.lfs.2020.118493] [PMID: 32979360]
[80]
Shetty AK. Mesenchymal stem cell infusion shows promise for combating coronavirus (COVID-19)-induced pneumonia. Aging Dis 2020; 11(2): 462-4.
[http://dx.doi.org/10.14336/AD.2020.0301] [PMID: 32257554]
[81]
Rajarshi K, Chatterjee A, Ray S. Combating COVID-19 with mesenchymal stem cell therapy. Biotechnol Reports 2020; 26: e00467.
[http://dx.doi.org/10.1016/j.btre.2020.e00467] [PMID: 32420049]
[82]
Karthik K, Senthilkumar TMA, Udhayavel S, Raj GD. Role of antibody-dependent enhancement (ADE) in the virulence of SARS-CoV-2 and its mitigation strategies for the development of vaccines and immunotherapies to counter COVID-19. Hum Vaccin Immunother 2020; 16(12): 3055-60.
[http://dx.doi.org/10.1080/21645515.2020.1796425] [PMID: 32845733]
[83]
Tan W, Lu Y, Zhang J, et al. Viral kinetics and antibody responses in patients with COVID-19. medRxiv 2020.
[http://dx.doi.org/10.1101/2020.03.24.20042382]
[84]
Saberian P, Falahi S, Baratloo A, et al. Changes in COVID-19 IgM and IgG antibodies in emergency medical technicians (EMTs). Am J Emerg Med 2021; 52: 59-63.
[http://dx.doi.org/10.1016/j.ajem.2021.11.019] [PMID: 34864629]
[85]
Moon C. Fighting COVID-19 exhausts T cells. Nat Rev Immunol 2020; 20(5): 277.
[http://dx.doi.org/10.1038/s41577-020-0304-7] [PMID: 32249845]
[86]
Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395(10223): 497-506.
[http://dx.doi.org/10.1016/S0140-6736(20)30183-5] [PMID: 31986264]
[87]
Fathi N, Rezaei N. Lymphopenia in COVID-19: Therapeutic opportunities. Cell Biol Int 2020; 44(9): 1792-7.
[http://dx.doi.org/10.1002/cbin.11403] [PMID: 32458561]
[88]
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.
[http://dx.doi.org/10.3389/fimmu.2020.00827] [PMID: 32425950]
[89]
Liu Y, Pang Y, Hu Z, et al. Thymosin alpha 1 (Tα1) reduces the mortality of severe COVID-19 by restoration of lymphocytopenia and reversion of exhausted T cells. Clin Infect Dis 2020; 71(16): 2150-7.
[PMID: 32442287]
[90]
Royer B, Kantelip J-P, Arock M. Modulation of interleukin-10 production by therapeutic drugs. Curr Med Chem Anti Inflamm Anti Allergy Agents 2003; 2(2): 95-106.
[http://dx.doi.org/10.2174/1568014033483815]
[91]
Liu Y, Sun W, Guo Y, et al. Association between platelet parameters and mortality in coronavirus disease 2019: Retrospective cohort study. Platelets 2020; 31(4): 490-6.
[http://dx.doi.org/10.1080/09537104.2020.1754383] [PMID: 32297540]
[92]
Osuchowski MF, Winkler MS, Skirecki T, et al. The COVID-19 puzzle: deciphering pathophysiology and phenotypes of a new disease entity. Lancet Respir Med 2021; 9(6): 622-42.
[http://dx.doi.org/10.1016/S2213-2600(21)00218-6] [PMID: 33965003]
[93]
Oudkerk M, Büller HR, Kuijpers D, et al. Diagnosis, prevention, and treatment of thromboembolic complications in COVID-19: Report of the national institute for public health of the netherlands. Radiology 2020; 297(1): E216-22.
[http://dx.doi.org/10.1148/radiol.2020201629] [PMID: 32324101]
[94]
Guglielmetti G, Quaglia M, Sainaghi PP, et al. “War to the knife” against thromboinflammation to protect endothelial function of COVID-19 patients. Crit Care 2020; 24(1): 365.
[http://dx.doi.org/10.1186/s13054-020-03060-9] [PMID: 32560665]
[95]
Connors JM, Levy JH. Thromboinflammation and the hypercoagulability of COVID-19. J Thromb Haemost 2020; 18(7): 1559-61.
[http://dx.doi.org/10.1111/jth.14849] [PMID: 32302453]
[96]
Hadid T, Kafri Z, Al-Katib A. Coagulation and anticoagulation in COVID-19. Blood Rev 2021; 47: 100761.
[http://dx.doi.org/10.1016/j.blre.2020.100761] [PMID: 33067035]
[97]
Caramaschi S, Kapp ME, Miller SE, et al. Histopathological findings and clinicopathologic correlation in COVID-19: A systematic review. Mod Pathol 2021; 34(9): 1614-33.
[http://dx.doi.org/10.1038/s41379-021-00814-w] [PMID: 34031537]
[98]
Rico-Mesa JS, Rosas D, Ahmadian-Tehrani A, White A, Anderson AS, Chilton R. The role of anticoagulation in COVID-19-induced hyper-coagulability. Curr Cardiol Rep 2020; 22(7): 53.
[http://dx.doi.org/10.1007/s11886-020-01328-8] [PMID: 32556892]
[99]
Thachil J. The versatile heparin in COVID-19. J Thromb Haemost 2020; 18(5): 1020-2.
[http://dx.doi.org/10.1111/jth.14821] [PMID: 32239799]
[100]
Rapkiewicz AV, Mai X, Carsons SE, et al. Megakaryocytes and platelet-fibrin thrombi characterize multi-organ thrombosis at autopsy in COVID-19: A case series. EClinicalMedicine 2020; 24: 100434.
[http://dx.doi.org/10.1016/j.eclinm.2020.100434] [PMID: 32766543]
[101]
Lax SF, Skok K, Zechner P, et al. Pulmonary arterial thrombosis in COVID-19 with fatal outcome: Results from a prospective, single-center, clinicopathologic case series. Ann Intern Med 2020; 173(5): 350-61.
[http://dx.doi.org/10.7326/M20-2566] [PMID: 32422076]
[102]
Tremblay D, van Gerwen M, Alsen M, et al. Impact of anticoagulation prior to COVID-19 infection: A propensity score-matched cohort study. Blood 2020; 136(1): 144-7.
[http://dx.doi.org/10.1182/blood.2020006941] [PMID: 32462179]
[103]
Falahi S, Kenarkoohi A. Host factors and vaccine efficacy: Implications for COVID-19 vaccines. J Med Virol 2021 jmv.27485.
[http://dx.doi.org/10.1002/jmv.27485] [PMID: 34845730]
[104]
de Loyola MB, Dos Reis TTA, de Oliveira GXLM, da Fonseca Palmeira J, Argañaraz GA, Argañaraz ER. Alpha-1-antitrypsin: A possible host protective factor against COVID-19. Rev Med Virol 2021; 31(2): e2157.
[http://dx.doi.org/10.1002/rmv.2157] [PMID: 32844538]
[105]
Oguntuyo KY, Stevens CS, Siddiquey MN, et al. In plain sight: The role of alpha-1-antitrypsin in COVID-19 pathogenesis and therapeu-tics. bioRxiv 2020; 2020.08.14.248880.
[http://dx.doi.org/10.1101/2020.08.14.248880]
[106]
Li X, Geng M, Peng Y, Meng L, Lu S. Molecular immune pathogenesis and diagnosis of COVID-19. J Pharm Anal 2020; 10(2): 102-8.
[http://dx.doi.org/10.1016/j.jpha.2020.03.001] [PMID: 32282863]
[107]
de Sousa E, Ligeiro D, Lérias JR, et al. Mortality in COVID-19 disease patients: Correlating the association of major histocompatibility complex (MHC) with severe acute respiratory syndrome 2 (SARS-CoV-2) variants. Int J Infect Dis 2020; 98: 454-9.
[http://dx.doi.org/10.1016/j.ijid.2020.07.016] [PMID: 32693089]
[108]
Meffre E, Iwasaki A. Interferon deficiency can lead to severe COVID. Nature 2020; 587(7834): 374-6.
[http://dx.doi.org/10.1038/d41586-020-03070-1] [PMID: 33139913]
[109]
van der Made CI, Simons A, Schuurs-Hoeijmakers J, et al. Presence of genetic variants among young men with severe COVID-19. JAMA 2020; 324(7): 663-73.
[http://dx.doi.org/10.1001/jama.2020.13719] [PMID: 32706371]
[110]
Hou Y, Zhao J, Martin W, et al. New insights into genetic susceptibility of COVID-19: An ACE2 and TMPRSS2 polymorphism analysis. BMC Med 2020; 18(1): 216.
[http://dx.doi.org/10.1186/s12916-020-01673-z] [PMID: 32664879]
[111]
Stawiski EW, Diwanji D, Suryamohan K, et al. Human ACE2 receptor polymorphisms predict SARS-CoV-2 susceptibility. bioRxiv 2020.
[http://dx.doi.org/10.1101/2020.04.07.024752]
[112]
Asselta R, Paraboschi EM, Mantovani A, Duga S. ACE2 and TMPRSS2 variants and expression as candidates to sex and country differ-ences in COVID-19 severity in Italy. Aging (Albany NY) 2020; 12(11): 10087-98.
[http://dx.doi.org/10.18632/aging.103415] [PMID: 32501810]
[113]
Zeberg H, Pääbo S. The major genetic risk factor for severe COVID-19 is inherited from Neanderthals. Nature 2020; 587(7835): 610-2.
[http://dx.doi.org/10.1038/s41586-020-2818-3] [PMID: 32998156]
[114]
Velavan TP, Pallerla SR, Rüter J, et al. Host genetic factors determining COVID-19 susceptibility and severity. EBioMedicine 2021; 72: 103629.
[http://dx.doi.org/10.1016/j.ebiom.2021.103629] [PMID: 34655949]
[115]
Thierry AR. Host/genetic factors associated with COVID-19 call for precision medicine. Precis Clin Med 2020; 3(3): 228-34.
[http://dx.doi.org/10.1093/pcmedi/pbaa026]
[116]
Ranjan J, Ravindra A, Mishra B. Gender and genetic factors impacting COVID-19 severity. J Family Med Prim Care 2021; 10(11): 3956-63.
[http://dx.doi.org/10.4103/jfmpc.jfmpc_769_21]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy