Acessibilidade / Reportar erro

COVID-19 and Acute Kidney Injury ― Direct and Indirect Pathophysiological Mechanisms Underlying Lesion Development

Abstract

COVID-19 is a pandemic disease caused by the SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) responsible for millions of deaths worldwide. Although the respiratory system is the main target of COVID-19, the disease can affect other organs, including the kidneys. Acute Kidney Injury (AKI), commonly seen in patients infected with COVID-19, has a multifactorial cause. Several studies associate this injury with the direct involvement of the virus in renal cells and the indirect damage stimulated by the infection. The direct cytopathic effects of SARS-CoV-2 are due to the entry and replication of the virus in renal cells, changing several regulatory pathways, especially the renin-angiotensin-aldosterone system (RAAS), with repercussions on the kallikrein-kinin system (KKS). Furthermore, the virus can deregulate the immune system, leading to an exaggerated response of inflammatory cells, characterizing the state of hypercytokinemia. The such exaggerated inflammatory response is commonly associated with hemodynamic changes, reduced renal perfusion, tissue hypoxia, generation of reactive oxygen species (ROS), endothelial damage, and coagulopathies, which can result in severe damage to the renal parenchyma. Thereby, understanding the molecular mechanisms and pathophysiology of kidney injuries induced by SARS-COV-2 is of fundamental importance to obtaining new therapeutic insights for the prevention and management of AKI.

Key words
Acute kidney injury; angiotensin; coronavirus; inflammation; SARS-CoV-2

INTRODUCTION

In December 2019, the attention of health authorities turned to COVID-19, a disease caused by the SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) responsible for millions of deaths worldwide (Chen et al. 2020CHEN G ET AL. 2020. Clinical and Immunological Features of Severe and Moderate Coronavirus Disease 2019. J Clin Investig 130(5): 2620-2629.). The clinical spectrum of the disease ranges from typical symptoms of upper respiratory tract infection to more severe complications, such as pneumonia and severe acute respiratory syndrome (SARS) (Qi et al. 2021QI K ET AL. 2021. Clinical, Laboratory, and Imaging Features of Pediatric COVID-19: A Systematic Review and Meta-Analysis. Medicine 100(15): e25230.). Although the respiratory system is the main target of COVID-19, the disease can also have repercussions on other organs, including the kidneys (Perico et al. 2020PERICO L, BENIGNI A & REMUZZI G. 2020. Should COVID-19 Concern Nephrologists? Why and to What Extent? The Emerging Impasse of Angiotensin Blockade. Nephron 144(5): 213-221., Adapa et al. 2020ADAPA S, CENNA A, BALLA M, MERUGU GP, KODURI NM, DAGGUBATI SR, GAYAM V, NARAMALA S & KONALA VM. 2020. COVID-19 Pandemic Causing Acute Kidney Injury and Impact on Patients With Chronic Kidney Disease and Renal Transplantation. J Clin Med Res 12(6): 352-361., El-Sayed et al. 2021EL-SAYED EE, ALLAYEH AK, SALEM AA, OMAR SM, ZAGHLOL SM, ABD-ELMAGUID HM, ABDUL-GHAFFAR MM & ELSHARKAWY MM. 2021. Incidence of Acute Kidney Injury among COVID-19 Patients in Egypt. Ren Replace Ther 7(1): 1-7.).

Studies investigating renal involvement in the course of COVID-19 disease are still limited. However, growing evidence indicates that renal changes are frequent and can range from mild proteinuria to more severe acute renal injury (AKI) (Li et al. 2020cLI Z ET AL. 2020d. Caution on Kidney Dysfunctions of COVID-19 Patients. medRxiv 2020.02.08.20021212. DOI: https://doi.org/10.1101/2020.02.08.20021212.
https://doi.org/10.1101/2020.02.08.20021...
, Adapa et al. 2020ADAPA S, CENNA A, BALLA M, MERUGU GP, KODURI NM, DAGGUBATI SR, GAYAM V, NARAMALA S & KONALA VM. 2020. COVID-19 Pandemic Causing Acute Kidney Injury and Impact on Patients With Chronic Kidney Disease and Renal Transplantation. J Clin Med Res 12(6): 352-361., El-Sayed et al. 2021EL-SAYED EE, ALLAYEH AK, SALEM AA, OMAR SM, ZAGHLOL SM, ABD-ELMAGUID HM, ABDUL-GHAFFAR MM & ELSHARKAWY MM. 2021. Incidence of Acute Kidney Injury among COVID-19 Patients in Egypt. Ren Replace Ther 7(1): 1-7., Almeida et al. 2021ALMEIDA DC ET AL. 2021. Acute Kidney Injury: Incidence, Risk Factors, and Outcomes in Severe COVID-19 Patients. PLoS ONE 16(5): e0251048.). AKI is a clinical syndrome characterized by a sudden decline in glomerular filtration rate (GFR), which may result in increased serum levels of nitrogen slag, changes in hydro electrolytic homeostasis, and acid-base imbalance (KDIGO 2012KDIGO - KIDNEY DISEASE: IMPROVING GLOBAL OUTCOMES. 2012. KDIGO Clinical Practice Guideline for Acute Kidney Injury. Kidney Int Suppl 2: 1-138.). Recently, several studies have demonstrated the occurrence of AKI in the course of the disease of the new Coronavirus (Zhou et al. 2020ZHOU F ET AL. 2020. Clinical Course and Risk Factors for Mortality of Adult Inpatients with COVID-19 in Wuhan, China: A Retrospective Cohort Study. Lancet 395(10229): 1054-1062. https://doi.org/10.1016/s0140-6736(20)30566-3.
https://doi.org/10.1016/s0140-6736(20)30...
, Li et al. 2020aLI G ET AL. 2020a. Coronavirus Infections and Immune Responses. J Med Virol 92 (4): 424-432., El-Sayed et al. 2021EL-SAYED EE, ALLAYEH AK, SALEM AA, OMAR SM, ZAGHLOL SM, ABD-ELMAGUID HM, ABDUL-GHAFFAR MM & ELSHARKAWY MM. 2021. Incidence of Acute Kidney Injury among COVID-19 Patients in Egypt. Ren Replace Ther 7(1): 1-7.). AKI is a prevalent complication among patients with COVID-19 and an indicator of disease severity, whose incidence ranges from 30% to 50% in patients hospitalized in the intensive care unit (ICU) (Ahmadian et al. 2020AHMADIAN E, KHATIBI SMH, SOOFIYANI SR, ABEDIAZAR S, SHOJA MM, ARDALAN M & VAHED SZ. 2020. Covid-19 and Kidney Injury: Pathophysiology and Molecular Mechanisms. Rev Med Virol 31(3): e2176. Accessed July 1, 2020. https://doi.org/10.1002/rmv.2176.
https://doi.org/10.1002/rmv.2176...
, Cheng et al. 2020bCHENG Y, LUO R, WANG K, ZHANG M, WANG Z, DONG L, LI J, YAO Y, GE S & XU G. 2020b. Kidney Impairment Is Associated with in-Hospital Death of COVID-19 Patients. DOI: https://doi.org/10.1101/2020.02.18.20023242.
https://doi.org/10.1101/2020.02.18.20023...
, Cummings et al. 2020CUMMINGS MJ ET AL. 2020. Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 in New York City: a prospective cohort study. Lancet 395(10239): 1763-1770. DOI: 10.1016/S0140-6736(20)31189-2.). Evidence also points out that chronic kidney disease (CKD) and elevated serum creatinine levels may represent a higher risk for developing more severe forms of COVID-19, with an important impact on the morbidity and mortality of hospitalized patients (Adapa et al. 2020ADAPA S, CENNA A, BALLA M, MERUGU GP, KODURI NM, DAGGUBATI SR, GAYAM V, NARAMALA S & KONALA VM. 2020. COVID-19 Pandemic Causing Acute Kidney Injury and Impact on Patients With Chronic Kidney Disease and Renal Transplantation. J Clin Med Res 12(6): 352-361.).

The underlying mechanisms of SARS-CoV-2-induced AKI are not yet fully understood. However, several factors may be involved in the pathogenesis of renal damage, including mainly local and systemic inflammatory responses, endothelial dysfunction, hypercoagulation, and imbalance of the renin-angiotensin-aldosterone system (RAAS) (Qu et al. 2014QU X, WANG C, ZHANG J, QIE G & ZHOU J. 2014. The Roles of CD147 And/or Cyclophilin A in Kidney Diseases. Mediators Inflamm 71(15): 762-768. DOI: https://doi.org/10.1155/2014/728673.
https://doi.org/10.1155/2014/728673...
, Li et al. 2020dLI Y, ZHOU W, YANG L & YOU R. 2020c. Physiological and Pathological Regulation of ACE2, the SARS-CoV-2 Receptor. Pharmacol Res 157: 104833., Santoriello et al. 2020SANTORIELLO D, KHAIRALLAH P, BOMBACK AS, XU K, KUDOSE S, BATAL I, BARASCH J, RADHAKRISHNAN J, D’AGATI V & MARKOWITZ G. 2020. Postmortem Kidney Pathology Findings in Patients with COVID-19. J Am Soc Nephrol 31(9): 2158-2167., Hamming et al. 2004HAMMING IWT, BULTHUIS MLC, LELY AT, NAVIS GJ & VAN GOOR H. 2004. Tissue Distribution of ACE2 Protein, the Functional Receptor for SARS Coronavirus. A First Step in Understanding SARS Pathogenesis. J Pathol 203(2): 631-637.). Although still controversial, evidence suggests that the activation of these pathophysiological mechanisms is due to direct viral infection, due to the renal tropism of the virus, and/or indirect factors widely present in critically ill patients who need ventilatory and hemodynamic support (Ames et al. 2019AMES MK, ATKINS CE & PITT B. 2019. The Renin-Angiotensin-Aldosterone System and Its Suppression. J Vet Intern Med 33(2): 363-382., Ronco & Reis 2020RONCO C & REIS T. 2020. Kidney Involvement in COVID-19 and Rationale for Extracorporeal Therapies. Nat Rev Nephrol 16(6): 308-310.). Thus, the current literature highlights two main hypotheses to explain AKI: the hypothesis of direct tissue injury and the hypothesis of indirect tissue injury.

The first hypothesis, or hypothesis of direct tissue injury, is based on the ability of the circulating virus to enter the renal tubular cells through the angiotensin-converting enzyme 2 receptor (ACE2) expressed on proximal tubule cells, which is the main portion affected in kidney injury. ACE2 is a zinc metalloprotease, which shares homology with the angiotensin-converting enzyme (ACE) in its catalytic domain and provides different critical functions in the RAAS (Donoghue et al. 2000DONOGHUE M ET AL. 2000. A Novel Angiotensin-Converting Enzyme-Related Carboxypeptidase (ACE2) Converts Angiotensin I to Angiotensin 1-9. Circ Res 87(5): E1-E9.). Thus, when the virus enters and multiplies in renal cells, several pathophysiological mechanisms are activated, including the main dysregulation of RAAS, kallikrein-kinin system (KKS), and inflammatory pathways which culminate in acute tissue injury (Chu et al. 2005CHU KH ET AL. 2005. Acute Renal Impairment in Coronavirus-Associated Severe Acute Respiratory Syndrome. Kidney Int 67(2): 698-705., Bradley et al. 2020BRADLEY BT ET AL. 2020. Histopathology and Ultrastructural Findings of Fatal COVID-19 Infections in Washington State: A Case Series. Lancet 396(10247): 320-332.). On the other hand, the second hypothesis is based on a series of systemic developments resulting from initial viral infection, including exacerbation of the immune response and circulatory and hemodynamic changes (Santoriello et al. 2020SANTORIELLO D, KHAIRALLAH P, BOMBACK AS, XU K, KUDOSE S, BATAL I, BARASCH J, RADHAKRISHNAN J, D’AGATI V & MARKOWITZ G. 2020. Postmortem Kidney Pathology Findings in Patients with COVID-19. J Am Soc Nephrol 31(9): 2158-2167., Qin et al. 2020QIN C ET AL. 2020. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infec Dis 71(15): 762-768.).

Given the clinical importance of the kidney repercussions associated with COVID-19 in hospitalized patients, as well as its impact on the quality of life of these patients after the hospitalization period, the elucidation of the pathophysiological mechanisms underlying SARS-CoV-2-induced AKI is essential to characterize the spectrum of lesions, as well as to define the appropriate and most effective therapeutic approach for each case. Therefore, the main objective of this article is to discuss current information on the pathophysiological aspects underlying SARS-CoV-2-induced AKI, exploring the direct and indirect molecular mechanisms involved in the development of lesions.

MATERIALS AND METHODS

We performed the bibliographic search in the MedLine/PubMed electronic databases. The initial literature screening was based on the title and abstract, using the following keywords: COVID-19, SARS-COV-2, acute kidney injury, renal, histopathology, pathophysiological mechanisms, and pathophysiology. Targeted searches were conducted using terms such as inflammation, complement system, RAAS, ACE2, kallikrein-kinin system, and coagulopathy. We made no restrictions on the publication dates of articles. The articles were independently selected by three authors, which included publications from 1998 to 2021.

PATHOPHYSIOLOGICAL MECHANISMS BASED ON THE HYPOTHESIS OF DIRECT TISSUE DAMAGE

Evidence of the presence of SARS-Cov2 in the renal cells

The hypothesis of direct tissue injury is based on the ability of SARS-CoV-2 to enter and replicate in the kidney cells, with consequent activation of several signaling pathways capable of deregulating cellular homeostasis, culminating in structural changes and loss of its functions (Ahmadian et al. 2020AHMADIAN E, KHATIBI SMH, SOOFIYANI SR, ABEDIAZAR S, SHOJA MM, ARDALAN M & VAHED SZ. 2020. Covid-19 and Kidney Injury: Pathophysiology and Molecular Mechanisms. Rev Med Virol 31(3): e2176. Accessed July 1, 2020. https://doi.org/10.1002/rmv.2176.
https://doi.org/10.1002/rmv.2176...
). This hypothesis has been supported by some studies that have demonstrated evidence of direct viral infection through the detection of SARS-CoV-2 mRNA and proteins in tubular and glomerular recurrent cells (Chu et al. 2005CHU KH ET AL. 2005. Acute Renal Impairment in Coronavirus-Associated Severe Acute Respiratory Syndrome. Kidney Int 67(2): 698-705., Ahmadian et al. 2020AHMADIAN E, KHATIBI SMH, SOOFIYANI SR, ABEDIAZAR S, SHOJA MM, ARDALAN M & VAHED SZ. 2020. Covid-19 and Kidney Injury: Pathophysiology and Molecular Mechanisms. Rev Med Virol 31(3): e2176. Accessed July 1, 2020. https://doi.org/10.1002/rmv.2176.
https://doi.org/10.1002/rmv.2176...
, Yin & Wunderink 2018YIN Y & WUNDERINK RG. 2018. MERS, SARS and Other Coronaviruses as Causes of Pneumonia. Respirol 23(2): 130-137. https://doi.org/10.1111/resp.13196.
https://doi.org/10.1111/resp.13196...
).

Histopathological and immunohistochemical analyses of renal tissue in patients with COVID-19 have contributed both to the characterization of lesions and the identification of antigenic particles in different segments of the nephron. In this sense, studies with transmission electron microscopy have identified the presence of virus-like particles in the cytoplasm of the proximal tubular epithelium, podocytes, and to a lesser extent in the distal tubules (Su et al. 2020SU H ET AL. 2020. Renal Histopathological Analysis of 26 Postmortem Findings of Patients with COVID-19 in China. Kidney Int 98(1): 219-227., Diao et al. 2021DIAO B ET AL. 2021. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Nat Comm 12(1): 2506., Werion et al. 2020WERION A ET AL. 2020. SARS-CoV-2 Causes a Specific Dysfunction of the Kidney Proximal Tubule. Kidney Int 98(5): 1296-1307., Bradley et al. 2020BRADLEY BT ET AL. 2020. Histopathology and Ultrastructural Findings of Fatal COVID-19 Infections in Washington State: A Case Series. Lancet 396(10247): 320-332.). Some studies have also demonstrated the presence of SARS-CoV-2 by immunohistochemical expression of viral nucleocapsid protein (NP) antigen in renal tubules (Diao et al. 2020DIAO B ET AL. 2020. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) medRxiv. 2020: 2020.03. 04.20031120, Szabolcs et al. 2021SZABOLCS M ET AL. 2021. Identification of Immunohistochemical Reagents for In Situ Protein Expression Analysis of Coronavirus-Associated Changes in Human Tissues. Appl Immunohistochem Mol Morphol 29(1): 5-12.). In addition, coronavirus-like particles were observed in vacuoles or cisterns of the rough endoplasmic reticulum (RER) and the smooth endoplasmic reticulum (REL) in cells of the proximal tubule by electron microscopy (Werion et al. 2020WERION A ET AL. 2020. SARS-CoV-2 Causes a Specific Dysfunction of the Kidney Proximal Tubule. Kidney Int 98(5): 1296-1307.). These findings agree with those reported by Diao et al. (2020)DIAO B ET AL. 2020. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) medRxiv. 2020: 2020.03. 04.20031120 who demonstrated that infected renal cells of the proximal tubules were quite swollen, with expansion and structural impairment of RER, REL, mitochondria, and lysosomes.

The presence of viral fragments in the urine is also a strong indication of direct infection of SARS-CoV-2 in the kidneys. In this sense, Caceres et al. (2021)CACERES PS ET AL. 2021. High SARS-CoV-2 Viral Load in Urine Sediment Correlates with Acute Kidney Injury and Poor COVID-19 Outcome. J Am Soc Nephrol 32(10): 2517-2528. observed that among 52 patients with COVID-19, 20 (39%) SARS-CoV-2 U-viral load, of which 17 developed AKI with an average U-viral load four-times higher than patients with COVID-19 who did not have AKI. The predilection of the virus for the kidneys can be partially explained by the fact that urine is an acidic environment, which allows the activation of the enzyme cysteine cathepsins, a primary protease pointed out to facilitate the entry of the virus into renal cells through endocytosis (Chong & Saha 2021WOON HC & SAHA BK. 2021. Relationship Between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and the Etiology of Acute Kidney Injury (AKI). Am J Med Sci 361 (3): 287-296.). On the other hand, some studies have not observed the presence of SARS-CoV-2 viral particles or RNA in the kidney of patients with COVID-19 (Bradley et al. 2020BRADLEY BT ET AL. 2020. Histopathology and Ultrastructural Findings of Fatal COVID-19 Infections in Washington State: A Case Series. Lancet 396(10247): 320-332., Wu et al. 2020WU H ET AL. 2020. AKI and Collapsing Glomerulopathy Associated with COVID-19 and High-Risk Genotype. J Am Soc Nephrol 31(8): 1688-1695., Sharma et al. 2020SHARMA P ET AL. 2020. COVID-19-Associated Kidney Injury: A Case Series of Kidney Biopsy Findings. J Am Soc Nephrol 31(9): 1948-1958., Kudose et al. 2020KUDOSE S ET AL. 2020. Kidney Biopsy Findings in Patients with COVID-19. J Am Soc Nephrol 31(9): 1959-1968.). However, it is not possible to completely rule out the presence of the virus in renal tissue in these studies due to the possibility that viral particles are in concentrations below the detection limit.

Therefore, collectively all these findings strongly suggest that COVID-19-induced AKI may involve direct infection of SARS-CoV-2 in the renal cells, triggering several mechanisms that culminate in the development of lesions in several parts of the nephron.

SARS-Cov-2 entry mechanisms in the cells

It is well established that SARS-CoV-2 uses the angiotensin-2-converter enzyme (ACE2) to enter host cells. In 2003 it was described as a receptor for the Severe Acute Respiratory Syndrome (SARS) coronavirus (Li et al. 2003LI W ET AL. 2003. Angiotensin-Converting Enzyme 2 Is a Functional Receptor for the SARS Coronavirus. Nature 426(6965): 450-454. DOI: https://doi.org/10.1038/nature02145.
https://doi.org/10.1038/nature02145...
). ACE2 is an insoluble transmembrane metalloprotease homologous to ACE, 42% sequence identity and 61% sequence similarity (Donoghue et al. 2000DONOGHUE M ET AL. 2000. A Novel Angiotensin-Converting Enzyme-Related Carboxypeptidase (ACE2) Converts Angiotensin I to Angiotensin 1-9. Circ Res 87(5): E1-E9.). Despite this, ACE2 contains a single zinc-binding domain HEXXH, which is homologous to the active sites of ACE, it is not inhibited by ACE inhibitors. This enzyme exists both as a membrane-associated form and as a secreted form, being widely distributed in cells of various body structures, such as the heart, lung, testicles, hypothalamus, and kidneys (Donoghue et al. 2000DONOGHUE M ET AL. 2000. A Novel Angiotensin-Converting Enzyme-Related Carboxypeptidase (ACE2) Converts Angiotensin I to Angiotensin 1-9. Circ Res 87(5): E1-E9., Li et al. 2020c). ACE2, in addition to playing a relevant role in the route of direct infection by SARS-CoV-2, is also part of a regulatory system called the renin-angiotensin-aldosterone system (RAAS). This system consists of a complex and elegant cascade of regulatory and counterregulatory vasoactive peptides responsible for controlling blood pressure, acting both systemically and locally in the kidneys to regulate vascular tone, renal sodium reabsorption, and extracellular fluid volume (Ames et al. 2019AMES MK, ATKINS CE & PITT B. 2019. The Renin-Angiotensin-Aldosterone System and Its Suppression. J Vet Intern Med 33(2): 363-382.). Furthermore, the deregulation of this system is widely associated with the progression of lesions, with induction of pro-inflammatory and profibrotic pathways in various types of renal diseases, including COVID-19-induced AKI (Chong & Saha 2021, Xu et al. 2017XU Z, LI W, HAN J, ZOU C, HUANG W, YU W, SHAN X, LUM H, LI X & LIANG G. 2017. Angiotensin II Induces Kidney Inflammatory Injury and Fibrosis through Binding to Myeloid Differentiation Protein-2 (MD2). Sci Rep 7(1): 1-11. https://doi.org/10.1038/srep44911.
https://doi.org/10.1038/srep44911...
). Although a broader approach to RAAS is beyond the scope of this article, Figure 1 provides an overview of the operation of the classical axis (vasoconstrictor) and the non-classical axis (vasorelaxant) of this system. Briefly, this system starts with the production of angiotensinogen in the liver, which will be cleaved into angiotensin (Ang) I by renin, an enzyme synthesized and released in the kidneys in response to stimuli related to low blood pressure. From then on, the system is divided into two distinct and antagonistic axes: the classic and non-classic axes (Povlsen et al. 2020POVLSEN AL, GRIMM D, WEHLAND M, INFANGER M & KRÜGER M. 2020. The Vasoactive Mas Receptor in Essential Hypertension. J Clin Med Res 9(1): 267. https://doi.org/10.3390/jcm9010267.
https://doi.org/10.3390/jcm9010267...
). The classic axis is described by the conversion of Ang I into Ang II, through ACE, and the consequent binding of this peptide to the type 1 Ang II receptor (AT1-R), leading mainly to vasoconstriction. On the other hand, the most important active peptide of the non-classical axis is Ang 1-7, which is generated from the hydrolysis of Ang II by the ACE2 action. ACE2 can also convert Ang I to Ang 1-9, which in turn is cleaved by neutral endopeptidase (NEP) or ACE to produce Ang 1-7. Finally, Ang I can also be cleaved to Ang 1-7 by the prolyl carboxypeptidase of Ang II or directly by the action of NEP. Thus, Ang 1-7 binds to its MAS receptor (MAS-R), producing, among other effects, increasing endothelial function, potent vasodilation, anti-cardiac hypertrophy, besides anti-fibrotic, anti-inflammatory, and anti-apoptotic effects (Ferreira & Santos 2005FERREIRA AJ & SANTOS RAS. 2005. Cardiovascular Actions of Angiotensin-(1-7). Rev Bras Pesqui Med Biol 38(4): 499-507., Jiang et al. 2014JIANG F, YANG J, ZHANG Y, DONG M, WANG S, ZHANG Q, LIU FF, ZHANG K & ZHANG C. 2014. Angiotensin-Converting Enzyme 2 and Angiotensin 1-7: Novel Therapeutic Targets. Nat Rev Cardiol 11(7): 413-426., Santos et al. 2003SANTOS RA ET AL. 2003. Angiotensin-(1-7) Is an Endogenous Ligand for the G Protein-Coupled Receptor Mas. Proc Nat Acad Sci USA 100(14): 8258-8263. DOI: https://doi.org/10.1073/pnas.1432869100.
https://doi.org/10.1073/pnas.1432869100...
).

Figure 1
Classical and non-classical axis of the renin angiotensin aldosterone system. Renin, produced in the kidneys, cleaves the angiotensinogen synthesized in the liver to form angiotensin (Ang) I, which will later be converted into Ang II through the action of the Angiotensin-Converting Enzyme (ACE). Other enzymes, including chymase and cathepsin, serve as an alternative pathway for converting Ang I to Ang II. Ang I also serves as a substrate for neprilysin (NEP) and prolyl endopeptidase (PE), which cleaves it to form Ang 1-7. Furthermore, PE is able to cleave Ang II into Ang 1-7. Another important enzyme for the functioning of this system is the ACE2, responsible for the transformation of Ang I into Ang 1-9 and Ang II into Ang 1-7. Ang II, in addition to stimulating the production of aldosterone in the adrenal gland, binds to its AT1 receptor (AT1R) to activate the classic axis of this system. On the other hand, Ang 1-7 binds to its MAS receptor (MASR), activating the non-classical axis, capable of neutralizing the vasoconstrictor axis.

Studies report that the SARS-CoV-2 virus has an envelope of glycoproteins, whose subunit S1 binds to the ACE2 receptor present on the cell surface, allowing its transport through the cytoplasmic membrane (Soleimani 2020SOLEIMANI M. 2020. Acute Kidney Injury in SARS-CoV-2 Infection: Direct Effect of Virus on Kidney Proximal Tubule Cells. Int J Mol Sci 21(9): 3275., Hoffmann et al. 2020HOFFMANN M ET AL. 2020 SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Cell 181(2): 271-280.). In the kidney, it was observed that the expression of the ACE2 receptor occurred sharply in the proximal tubules, followed by a smaller extent in the podocytes, and rarely found in the glomeruli (Mizuiri & Ohashi 2015MIZUIRI S & OHASHI Y. 2015. ACE and ACE2 in Kidney Disease. World J Nephrol 4(1): 74-82., Perico et al. 2020PERICO L, BENIGNI A & REMUZZI G. 2020. Should COVID-19 Concern Nephrologists? Why and to What Extent? The Emerging Impasse of Angiotensin Blockade. Nephron 144(5): 213-221., Hamming et al. 2004HAMMING IWT, BULTHUIS MLC, LELY AT, NAVIS GJ & VAN GOOR H. 2004. Tissue Distribution of ACE2 Protein, the Functional Receptor for SARS Coronavirus. A First Step in Understanding SARS Pathogenesis. J Pathol 203(2): 631-637., Pan et al. 2020PAN XW, XU D, ZHANG H, ZHOU W, WANG LH & CUI XG. 2020. 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 46(6): 1114-1116.). In one of the first histopathological analyses of renal tissue in patients with AKI and positive for COVID-19, it was found that the SARS-CoV-2 ACE2 receptor was upregulated in the proximal tubular cells, especially in areas with an acute injury (Su et al. 2020SU H ET AL. 2020. Renal Histopathological Analysis of 26 Postmortem Findings of Patients with COVID-19 in China. Kidney Int 98(1): 219-227.). It is noteworthy to note that this increase was not related to therapeutic measures, since, when necessary, these patients were treated with calcium channel blockers for hypertension, while ACE inhibitors or angiotensin receptor blockers, indicated as supposedly responsible for the overregulation of these receptors, were not used (Su et al. 2020SU H ET AL. 2020. Renal Histopathological Analysis of 26 Postmortem Findings of Patients with COVID-19 in China. Kidney Int 98(1): 219-227.).

In addition to the ACE2 receptor, auxiliary proteins belonging to protease families, such as transmembrane serine protease 2 (TMPRSS2), act as co-receptors in collaboration with ACE2, cleaving the viral S protein and thus facilitating the entry of coronavirus into target cells (Soleimani 2020SOLEIMANI M. 2020. Acute Kidney Injury in SARS-CoV-2 Infection: Direct Effect of Virus on Kidney Proximal Tubule Cells. Int J Mol Sci 21(9): 3275., Hoffmann et al. 2020HOFFMANN M ET AL. 2020 SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Cell 181(2): 271-280., Zhou et al. 2015ZHOU Y ET AL. 2015 Protease Inhibitors Targeting Coronavirus and Filovirus Entry. Antiviral Res 116(April): 76-84.). Unlike airway epithelial cells, studies report that tubular cells express very low levels of TMPRSS2 (Soleimani 2020SOLEIMANI M. 2020. Acute Kidney Injury in SARS-CoV-2 Infection: Direct Effect of Virus on Kidney Proximal Tubule Cells. Int J Mol Sci 21(9): 3275., Vaarala et al. 2001VAARALA MH, PORVARI KS, KELLOKUMPU S, KYLLÖNEN AP & VIHKO PT. 2001. Expression of Transmembrane Serine Protease TMPRSS2 in Mouse and Human Tissues. J Pathol 193(1): 134-140. DOI: https://doi.org/10.1002/1096-9896(2000)9999:9999<::AID-PATH743>3.0.CO;2-T.
https://doi.org/10.1002/1096-9896(2000)9...
, Lucas et al. 2008LUCAS JM, TRUE L, HAWLEY S, MATSUMURA M, MORRISSEY C, VESSELLA R & NELSON PS. 2008. The Androgen-Regulated Type II Serine Protease TMPRSS2 Is Differentially Expressed and Mislocalized in Prostate Adenocarcinoma. J Pathol 215(2): 118-125. https://doi.org/10.1002/path.2330.
https://doi.org/10.1002/path.2330...
). However, these cells express abundant levels of primary protease potentials that can replace TMPRSS2, including cathepsin cysteine protease B/L, glutamyl aminopeptidase, and protease serine dipeptidyl peptidase 4 (DPP4) (Soleimani 2020SOLEIMANI M. 2020. Acute Kidney Injury in SARS-CoV-2 Infection: Direct Effect of Virus on Kidney Proximal Tubule Cells. Int J Mol Sci 21(9): 3275., Nistala & Savin 2017NISTALA R & SAVIN V. 2017. Diabetes, Hypertension, and Chronic Kidney Disease Progression: Role of DPP4. Am J Physiol Renal Physiol 312(4): F661-F670. DOI: https://doi.org/10.1152/ajprenal.00316.2016.
https://doi.org/10.1152/ajprenal.00316.2...
, Girardi et al. 2001GIRARDI ACC, DEGRAY BC, NAGY T, BIEMESDERFER D & ARONSON PS. 2001. Association of Na+-H+ Exchanger Isoform NHE3 and Dipeptidyl Peptidase IV in the Renal Proximal Tubule. J Biol Chem 276(49): 46671-46677., Qi et al. 2020QI F, QIAN S, ZHANG S & ZHANG Z. 2020. Single Cell RNA Sequencing of 13 Human Tissues Identify Cell Types and Receptors of Human Coronaviruses. Biochem Biophys Res Commun 526(1): 135-140.), thus facilitating the entry of SARS-CoV-2, especially into proximal tubule cells.

Another possible input route of SARS-CoV-2 into the cell is via CD147/basigin. CD147 is a transmembrane glycoprotein belonging to the immunoglobulin family, also known as extracellular matrix metalloproteinase inducer (EMMPRIN), which interacts with several other proteins, including caveolin-1, integrins, and cyclophilins, the latter having an important role in the coronavirus replication process (Wang et al. 2020WANG KE ET AL. 2020. SARS-CoV-2 Invades Host Cells via a Novel Route: CD147-Spike Protein. Biorxiv. https://doi.org/10.1101/2020.03.14.988345.
https://doi.org/10.1101/2020.03.14.98834...
). In normal kidneys, CD147 is highly expressed only on the basolateral side of tubular epithelial cells (Kosugi et al. 2015KOSUGI T, MAEDA K, SATO W, MARUYAMA S & KADOMATSU K. 2015. CD147 (EMMPRIN/Basigin) in Kidney Diseases: From an Inflammation and Immune System Viewpoint. Nephrol Dial Transplant 30(7): 1097-1103.). However, the expression of CD147 was also observed in interfacial and apical sides and in some podocytes and parietal cells in the kidneys of patients with COVID-19 (Su et al. 2021SU H, WAN C, WANG ZD, GAO Y, LI YC, TANG F, ZHU HY, YI LX & ZHANG C. 2021. Expression of CD147 and Cyclophilin A in Kidneys of Patients with COVID-19. Clin J Am Soc Nephrol 16(4): 618-619.). Therefore, all this evidence suggests that after causing the lung infection, the virus can enter the bloodstream, accumulate in the kidneys and cause damage to kidney cells. Figure 2 summarizes the possibilities of entry of SARS-CoV-2 into the host cell, as well as the interaction between the receptor and the cell co-receptor with the virus spike protein.

Figure 2
Direct effects of SARS-CoV-2 associated with acute kidney injury. SARS-CoV-2 invades the host cell through interaction with ACE2 present in the cell membrane. For this, the presence of co-receptors that contribute to the spike protein cleavage and subsequent virus internalization is necessary. Among the possible auxiliary proteins, TMPRSS2, cysteine cathepsin B/L, glutamyl aminopeptidase, and serine protease dipeptidyl peptidase 4 (DPP4) stand out. Virus entry can also occur through CD147, a glycoprotein present on the basolateral and apical face of cells from patients with COVID-19. Down-regulation of ACE2 stimulates hyperactivation of the classical RAAS axis, as well as the DABK-BKB1R axis of the kallikrein-kinin system. CD147, in addition to facilitating the entry of the virus, induces the expression of inflammatory cytokines. These events contribute to viral pathogenesis by aggravating kidney damage.

Direct mechanisms of tissue injury by SARS-CoV-2

The pathophysiological mechanisms underlying direct lesions induced by SARS-Cov-2 are not yet fully understood. However, evidence suggests that after the entry of the virus into the kidney cells, the main mechanisms of injury include the deregulation of RAAS and the kallikrein-kinin system (KKS), and the escalation of the inflammatory process, vascular permeability, vasoconstriction, and production of reactive oxygen species (ROS) (Figure 2).

As mentioned, ACE2 is very important for the balance of RAAS axes through the production of Ang (1-7), whose biochemical and functional features promote several cytoprotective effects that counter-regulate the effects of Ang II, including potent vasodilator and antiproliferative effects (Ferreira & Santos 2005FERREIRA AJ & SANTOS RAS. 2005. Cardiovascular Actions of Angiotensin-(1-7). Rev Bras Pesqui Med Biol 38(4): 499-507.). Furthermore, Ang (1-7) confers a renoprotective effect reducing the inflammatory process, vasoconstriction, and thrombotic events (Mizuiri & Ohashi 2015MIZUIRI S & OHASHI Y. 2015. ACE and ACE2 in Kidney Disease. World J Nephrol 4(1): 74-82.). However, when using the ACE2 receptors to enter the cell, SARS-CoV-2 negatively regulates its expression either by sequestration and internalization or by cleavage of its extracellular domain (Nicolau et al. 2020NICOLAU LAD, MAGALHÃES PJC & VALE ML. 2020. What Would Sérgio Ferreira Say to Your Physician in This War against COVID-19: How about Kallikrein/kinin System? Med Hypotheses 143: 109886.). Thus, the pathological process mediated by COVID-19 is associated with the deregulation of RAAS due to excessive concentrations of Ang-II in circulation and increased activation of the AT1 receptor, resulting in the exacerbated activation of its classical axis. The Ang-II surplus, in turn, promotes a decrease in renal blood flow through its vasoconstrictive property, besides enabling the increase of the inflammatory process, cell proliferation, and generation of ROS (Mizuiri & Ohashi 2015MIZUIRI S & OHASHI Y. 2015. ACE and ACE2 in Kidney Disease. World J Nephrol 4(1): 74-82.). Therefore, it is possible that patients diagnosed with COVID-19 may develop AKI due to ischemia secondary to vasoconstriction and decreased renal perfusion (Chong & Saha 2021, Ng et al. 2020NG JH, BIJOL V, SPARKS MA, SISE ME, IZZEDINE H & JHAVERI KD. 2020. Pathophysiology and Pathology of Acute Kidney Injury in Patients With COVID-19. Adv Chronic Kidney Dis 27(5): 365-376.).

Additionally, the lower expression of ACE2 receptors in the renal endothelium also generates disorders in the KKS, which further aggravates the scenario of tissue injury. This system is involved in inflammatory processes, control of vascular tone, electrolyte transport, and cell proliferation (Pesquero & Bader 1998PESQUERO JB & BADER M. 1998. Molecular Biology of the Kallikrein-Kinin System: From Structure to Function. Rev Med Pesqui Med Biol 31(9): 1197-1203.). Normally, kallikrein is activated by the Hageman factor in tissue injury situations and, once active, converts kininogen into kinins, among which stands out bradykinin, a peptide responsible for vasodilation and increased capillary permeability (Ramalho 2000RAMALHO FS. 2000. A Regeneração Hepática E Os Inibidores Da Enzima Conversora Da Angiotensina. Acta Cir Bras. DOI: https://doi.org/10.1590/s0102-86502000000600004.
https://doi.org/10.1590/s0102-8650200000...
). Bradykinin is rapidly cleaved by ACE in des-arg-bradykinin (DABK), a pro-inflammatory metabolite, which in turn is hydrolyzed by the action of ACE2 (Ramalho 2000RAMALHO FS. 2000. A Regeneração Hepática E Os Inibidores Da Enzima Conversora Da Angiotensina. Acta Cir Bras. DOI: https://doi.org/10.1590/s0102-86502000000600004.
https://doi.org/10.1590/s0102-8650200000...
, Sodhi et al. 2018SODHI CP, WOHLFORD-LENANE C, YAMAGUCHI Y, PRINDLE T, FULTON WB, WANG S, MCCRAY JR PB, CHAPPELL M, HACKAM DJ & JIA H. 2018. Biomarkers in Lung Diseases: From Pathogenesis to Prediction to New Therapies: Attenuation of Pulmonary ACE2 Activity Impairs Inactivation of Des-Arg9 bradykinin/BKB1R Axis and Facilitates LPS-Induced Neutrophil Infiltration. Am J Physiol Lung Cell Mol Physiol 314(1): L17-L31.). It is assumed that the downregulation of ACE2, induced by SARS-CoV-2 in the process of cell infection, results in the accumulation of DABK in the tissue, activating pro-inflammatory pathways that aggravate the damage of the renal parenchyma, as it occurs in the lungs (Mahmudpour et al. 2020MAHMUDPOUR M, ROOZBEH J, KESHAVARZ M, FARROKHI S & NABIPOUR I. 2020. COVID-19 Cytokine Storm: The Anger of Inflammation. Cytokine 133: 155151. https://doi.org/10.1016/j.cyto.2020.155151.
https://doi.org/10.1016/j.cyto.2020.1551...
, Nicolau et al. 2020NICOLAU LAD, MAGALHÃES PJC & VALE ML. 2020. What Would Sérgio Ferreira Say to Your Physician in This War against COVID-19: How about Kallikrein/kinin System? Med Hypotheses 143: 109886.). ACE2 proteolytic activity is inhibited after coronavirus cell infection, which turns it unable to inactivate DABK. As described by Oliveira et al. (2021)OLIVEIRA LCG, CRUZ NAN, RICELLI B, TEDESCO-SILVA H, MEDINA-PESTANA JO & CASARINI DE. 2021. Interactions amongst Inflammation, Renin-Angiotensin-Aldosterone and Kallikrein-Kinin Systems: Suggestive Approaches for COVID-19 Therapy. J Venom Anim Toxins Incl Trop Dis 27(December). DOI: https://doi.org/10.1590/1678-9199-JVATITD-2020-0181.
https://doi.org/10.1590/1678-9199-JVATIT...
, under this condition, the lung environment is prone to kinin-dependent local vascular leakage leading to angioedema via B1R and eventually B2R. Also, in that study, it was described that this blockade leads to increased Ang II levels, which stimulates the non-blocked angiotensin II receptor, AT2R, and triggers intracellular acidification by inhibiting the amiloride-sensitive Na+/H+ exchanger (Oliveira et al. 2021OLIVEIRA LCG, CRUZ NAN, RICELLI B, TEDESCO-SILVA H, MEDINA-PESTANA JO & CASARINI DE. 2021. Interactions amongst Inflammation, Renin-Angiotensin-Aldosterone and Kallikrein-Kinin Systems: Suggestive Approaches for COVID-19 Therapy. J Venom Anim Toxins Incl Trop Dis 27(December). DOI: https://doi.org/10.1590/1678-9199-JVATITD-2020-0181.
https://doi.org/10.1590/1678-9199-JVATIT...
). In addition, stimulation of the AT2R by Ang II causes intracellular acidification that increases kininogenase activity by kallikrein activation, resulting in the kallikrein-kinin system stimulation via BK releasing (Tsutsumi et al. 1999TSUTSUMI Y ET AL. 1999. Angiotensin II Type 2 Receptor Overexpression Activates the Vascular Kinin System and Causes Vasodilation. J Clin Invest 104(7): 925-935. DOI: https://doi.org/10.1172/JCI7886.
https://doi.org/10.1172/JCI7886...
).

The local inflammatory process following the entry of SARS-CoV-2 into the cell can also be mediated through CD147 (Sodhi et al. 2018SODHI CP, WOHLFORD-LENANE C, YAMAGUCHI Y, PRINDLE T, FULTON WB, WANG S, MCCRAY JR PB, CHAPPELL M, HACKAM DJ & JIA H. 2018. Biomarkers in Lung Diseases: From Pathogenesis to Prediction to New Therapies: Attenuation of Pulmonary ACE2 Activity Impairs Inactivation of Des-Arg9 bradykinin/BKB1R Axis and Facilitates LPS-Induced Neutrophil Infiltration. Am J Physiol Lung Cell Mol Physiol 314(1): L17-L31.). As stated earlier, this receptor has interaction with several proteins, such as cyclophilins, caveolin-1, and integrins (Chueh et al. 2020CHUEH TI, ZHENG CM, HOU YC & LU KC. 2020. Novel Evidence of Acute Kidney Injury in COVID-19. J Clin Med Res 9(11): 3547. DOI: https://doi.org/10.3390/jcm9113547.
https://doi.org/10.3390/jcm9113547...
, Qu et al. 2014QU X, WANG C, ZHANG J, QIE G & ZHOU J. 2014. The Roles of CD147 And/or Cyclophilin A in Kidney Diseases. Mediators Inflamm 71(15): 762-768. DOI: https://doi.org/10.1155/2014/728673.
https://doi.org/10.1155/2014/728673...
). Among the proteins that interact with CD147, cyclophilin has been the target of several studies in recent years, especially the A cyclophilin subtype (CyPA) (Liu et al. 2013LIU X, ZHAO Z & LIU W. 2013. Insights into the Roles of Cyclophilin A during Influenza Virus Infection. Viruses 5(1): 182-191. DOI: https://doi.org/10.3390/v5010182.
https://doi.org/10.3390/v5010182...
, Tian et al. 2020TIAN L, LIU W & SUN L. 2020. Role of Cyclophilin A during Coronavirus Replication and the Antiviral Activities of Its Inhibitors. Chin J Biotechnol 36(4): 605-611. DOI: https://doi.org/10.13345/j.cjb.200049.
https://doi.org/10.13345/j.cjb.200049...
, Li et al. 2020bLI T, QUAN H, ZHANG H, LIN L, OU Q & CHEN K. 2020b. Silencing Cyclophilin A Improves Insulin Secretion, Reduces Cell Apoptosis, and Alleviates Inflammation as Well as Oxidant Stress in High Glucose-Induced Pancreatic β-Cells via MAPK/NF-Kb Signaling Pathway. Bioengineered 11(1): 1047-1057. DOI: https://doi.org/10.1080/21655979.2020.1823729., Liao et al. 2021LIAO Y, LUO D, PENG K & ZENG Y. 2021. Cyclophilin A: A Key Player for Etiological Agent Infection. Appl Microbiol Biotechnol 105(4): 1365-1377. DOI: https://doi.org/10.1007/s00253-021-11115-2.
https://doi.org/10.1007/s00253-021-11115...
). Although CyPA is found mainly in the intracellular environment, it can be secreted in the extracellular medium due to inflammatory stimuli. Its extracellular form interacts with the CD147 receptor performing its chemotactic activities for monocytes, neutrophils, and eosinophils (Qu et al. 2014QU X, WANG C, ZHANG J, QIE G & ZHOU J. 2014. The Roles of CD147 And/or Cyclophilin A in Kidney Diseases. Mediators Inflamm 71(15): 762-768. DOI: https://doi.org/10.1155/2014/728673.
https://doi.org/10.1155/2014/728673...
). Thus, from immunostaining of renal tissue samples biopsied from patients with SARS-CoV-2, Su et al. (2021)SU H, WAN C, WANG ZD, GAO Y, LI YC, TANG F, ZHU HY, YI LX & ZHANG C. 2021. Expression of CD147 and Cyclophilin A in Kidneys of Patients with COVID-19. Clin J Am Soc Nephrol 16(4): 618-619. observed increased CyPA expression in the tubular epithelium, as well as in podocytes and parietal cells, which may partially explain the inflammatory process present in the renal tissue of these patients. On the other hand, it is known that CyPA plays an important suppressive role in the development of TCD4+ cells, so the positive regulation of these primary proteases may be related to the records of lymphocytopenia observed in patients with COVID-19 (Su et al. 2021SU H, WAN C, WANG ZD, GAO Y, LI YC, TANG F, ZHU HY, YI LX & ZHANG C. 2021. Expression of CD147 and Cyclophilin A in Kidneys of Patients with COVID-19. Clin J Am Soc Nephrol 16(4): 618-619., Guan et al. 2020GUAN WJ ET AL. 2020. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 382(18): 1708-1720. DOI: https://doi.org/10.1056/NEJMoa2002032.
https://doi.org/10.1056/NEJMoa2002032...
).

Finally, CD147 involvement was also demonstrated in inflammatory processes in experimental AKI models. Kato et al. (2009)KATO N, YUZAWA Y, KOSUGI T, HOBO A, SATO W, MIWA Y, SAKAMOTO K, MATSUO S & KADOMATSU K. 2009. The E-Selectin Ligand Basigin/CD147 Is Responsible for Neutrophil Recruitment in Renal Ischemia/Reperfusion. J Am Soc Nephrol 20(7): 1565-1576. demonstrated that CD147 was associated with massive recruitment and infiltration of inflammatory cells in the renal tissue of CD147-positive mice with ischemia/reperfusion-induced AKI. However, when the mice CD147 deficient were analyzed, there was a decrease in the recruitment and infiltration of inflammatory cells in the tubular interstitium and greater preservation of renal function of these animals (Kato et al. 2009KATO N, YUZAWA Y, KOSUGI T, HOBO A, SATO W, MIWA Y, SAKAMOTO K, MATSUO S & KADOMATSU K. 2009. The E-Selectin Ligand Basigin/CD147 Is Responsible for Neutrophil Recruitment in Renal Ischemia/Reperfusion. J Am Soc Nephrol 20(7): 1565-1576.).

PATHOPHYSIOLOGICAL MECHANISMS BASED ON THE HYPOTHESIS OF INDIRECT TISSUE DAMAGE

In addition to the damage caused by the direct cytopathic effects of SARS-CoV-2 under the host renal cells, the virus can also induce an exaggerated systemic immune response responsible for several pathophysiological changes, including hemodynamic changes, generation of ROS, and coagulopathies, which culminate in renal parenchyma involvement, endothelial damage, and serious repercussions to renal function. The indirect mechanisms of renal injury induced by COVID-19 are represented in Figure 3.

Figure 3
Indirect effects of SARS-CoV-2 associated with acute kidney injury. The hypercytokinemia contributes to the dysregulation of the coagulation pathways, complement system, increased endothelial permeability, and generation of reactive oxygen species (ROS). An erythrocyte aggregation can be noted, obstructing the capillary lumen, contributing to renal hypoperfusion. Furthermore, sites with low oxygen concentrations can contribute to the generation of ROS, further enhancing the reduction in arteriolar blood flow. Increased vascular permeability is associated with the inflammatory response, facilitating the passage of immune cells and water to the interstitial space, with the consequent formation of edema. SARS-CoV-2, ROS, leukocytes, and the deposition of the complement system (C5b-9) can potentiate damage to kidney cells. The reduction in renal perfusion, together with direct damage to the cells of the proximal tubule, allows for acute tubular necrosis (ATN) and loss of the brush border, progressing to AKI.

Cytokine storm syndrome

The inflammatory process associated with SARS-CoV-2 initially occurs in the lungs, as it is well known (Brandão et al. 2020BRANDÃO SCS, GODOI ETAM, RAMOS JOX, MELO LMMP & SARINHO ESC. 2020. Severe COVID-19: Understanding the Role of Immunity, Endothelium, and Coagulation in Clinical Practice. J Vasc Bras 19: e20200131.). Then, at the systemic level, the immune response is responsible for neutralizing the viral replication process, limiting the spread of the virus, and performing the cleansing of infected cells (Ivashkiv & Donlin 2014LIONEL BI & DONLIN LT. 2014. Regulation of Type I Interferon Responses. Nat Rev Immunol 14(1): 36-49., Li et al. 2020aLI G ET AL. 2020a. Coronavirus Infections and Immune Responses. J Med Virol 92 (4): 424-432.). However, when super-activated, this response can generate a condition of widespread aggressive inflammation called hypercytokinemia or ‘cytokine storm’ (Noris et al. 2020NORIS M, BENIGNI A & REMUZZI G. 2020. The Case of Complement Activation in COVID-19 Multiorgan Impact. Kidney Int 98(2): 314-322.). This cytokine storm generates especially damage to the pulmonary parenchyma, but also serious hemodynamic and circulatory repercussions capable of promoting extensive renal involvement (Chueh et al. 2020CHUEH TI, ZHENG CM, HOU YC & LU KC. 2020. Novel Evidence of Acute Kidney Injury in COVID-19. J Clin Med Res 9(11): 3547. DOI: https://doi.org/10.3390/jcm9113547.
https://doi.org/10.3390/jcm9113547...
).

The exact mechanisms related to the outbreak of hypercytokinemia are not yet fully understood. However, it is known that some infections caused by viruses of the coronavirus family use mechanisms that control the host’s immune response, delaying the initial response against infection (Totura & Baric 2012TOTURA AL & BARIC RS. 2012. SARS Coronavirus Pathogenesis: Host Innate Immune Responses and Viral Antagonism of Interferon. Curr Opin Virol 2(3): 264., Channappanavar et al. 2019CHANNAPPANAVAR R ET AL. 2019. IFN-I Response Timing Relative to Virus Replication Determines MERS Coronavirus Infection Outcomes. J Clin Investig 129(9): 3625.). Such regulation can be observed with interferon (IFN), one of those responsible for the first line of defense against viral infections, through which the coronavirus induces a late response, making it difficult to clear viral in the body (Totura & Baric 2012TOTURA AL & BARIC RS. 2012. SARS Coronavirus Pathogenesis: Host Innate Immune Responses and Viral Antagonism of Interferon. Curr Opin Virol 2(3): 264., Channappanavar et al. 2019CHANNAPPANAVAR R ET AL. 2019. IFN-I Response Timing Relative to Virus Replication Determines MERS Coronavirus Infection Outcomes. J Clin Investig 129(9): 3625.). This late signaling of type I interferon (IFN-I) promotes the accumulation of macrophage-monocytes, resulting in high levels of pro-inflammatory cytokines and chemokines, which characterize the subsequent exacerbated immune response (Channappanavar et al. 2016CHANNAPPANAVAR R, FEHR AR, VIJAY R, MACK M, ZHAO J, MEYERHOLZ DK & PERLMAN S. 2016. Dysregulated Type I Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice. Cell Host Microbe 19(2): 181-193.). Evidence indicates that this cytokine storm may be related to the severity, morbidity and mortality of coronavirus. In this sense, Chen et al. (2020)CHEN G ET AL. 2020. Clinical and Immunological Features of Severe and Moderate Coronavirus Disease 2019. J Clin Investig 130(5): 2620-2629. evaluated the plasma levels of cytokines in patients infected with the new coronavirus, finding that the levels of interleukin 2R (IL-2R), IL-6, IL-10, and tumor necrosis factor α (TNF-α) were markedly higher in severe cases than in moderate cases. Overall, pro-inflammatory cytokines related to macrophages, particularly IL-6, IL-10, and TNF-α, were found to have increased significantly in most severe cases (Chen et al. 2020CHEN G ET AL. 2020. Clinical and Immunological Features of Severe and Moderate Coronavirus Disease 2019. J Clin Investig 130(5): 2620-2629.).

Another possible factor contributing to SARS-CoV-2-induced hypercytokinemia is lymphopenia. In this sense, it was demonstrated that patients with severe COVID-19 had a deeper reduction in the absolute number of total T lymphocytes, CD4, and CD8 T cells (Chen et al. 2020CHEN G ET AL. 2020. Clinical and Immunological Features of Severe and Moderate Coronavirus Disease 2019. J Clin Investig 130(5): 2620-2629.). Other observational studies have also reported lymphopenia as an important marker of immune system disorder, being present in most patients with COVID-19 during hospital admission (Qin et al. 2020QIN C ET AL. 2020. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infec Dis 71(15): 762-768., Huang et al. 2020HUANG C ET AL. 2020. Clinical Features of Patients Infected with 2019 Novel Coronavirus in Wuhan, China. Lancet 395(10223): 497-506., Guan et al. 2020GUAN WJ ET AL. 2020. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 382(18): 1708-1720. DOI: https://doi.org/10.1056/NEJMoa2002032.
https://doi.org/10.1056/NEJMoa2002032...
). The mechanism underlying lymphocyte depletion is still poorly understood, but some studies attribute an important role to cytokines, claiming that pro-inflammatory cytokines derived from IFN-α/β or mononuclear macrophages induce T-cell apoptosis, which further hinders viral clearance and contributes to the extent of the inflammatory process (Ye et al. 2020YE Q, WANG B & MAO J. 2020. The Pathogenesis and Treatment of the “Cytokine Storm” in COVID-19. J Infect 80(6): 607-613.). Another possible explanation would be direct infection of SARS-CoV-2 in lymphocytes since these cells express ACE2, which would allow the entry of the virus leading to activation and consequent cell death (Batlle et al. 2020BATLLE D, SOLER MJ, SPARKS MA, HIREMATH S, SOUTH AM, WELLING PA & SWAMINATHAN S. 2020. Acute Kidney Injury in COVID-19: Emerging Evidence of a Distinct Pathophysiology. J Am Soc Nephrol 31(7): 1380-1383. https://doi.org/10.1681/ASN.2020040419.
https://doi.org/10.1681/ASN.2020040419...
).

Once installed, the storm of cytokines can trigger manifestations in different organs, including the kidneys. Studies have shown that hypercytokinemia may have contributed to the appearance of structural damage and changes in renal function through coagulation pathway disorders, complement system performance and dysregulation, and exaggerated recruitment of pro-inflammatory cells (Gómez-Escobar et al. 2021GÓMEZ-ESCOBAR LG ET AL. 2021. Cytokine Signatures of End Organ Injury in COVID-19. Sci Rep 11(1): 1-15., Diao et al. 2020DIAO B ET AL. 2020. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) medRxiv. 2020: 2020.03. 04.20031120). Such alterations, in turn, are often associated with increased vascular permeability and the establishment of hemodynamic changes, triggering the reduction of renal perfusion and consequent acute tubular necrosis (ATN). Thus, ATN has been the main pathological change observed in patients with COVID-19 who developed AKI (Santoriello et al. 2020SANTORIELLO D, KHAIRALLAH P, BOMBACK AS, XU K, KUDOSE S, BATAL I, BARASCH J, RADHAKRISHNAN J, D’AGATI V & MARKOWITZ G. 2020. Postmortem Kidney Pathology Findings in Patients with COVID-19. J Am Soc Nephrol 31(9): 2158-2167., Golmai et al. 2020GOLMAI P, LARSEN CP, DEVITA MV, WAHL JS, WEINS A, RENNKE HG, BIJOL V & ROSENSTOCK JL. 2020. Histopathologic and Ultrastructural Findings in Postmortem Kidney Biopsy Material in 12 Patients with AKI and COVID-19. J Am Soc Nephrol 31(9): 1944-1947., Volbeda et al. 2021VOLBEDA M ET AL. 2021. Comparison of Renal Histopathology and Gene Expression Profiles between Severe COVID-19 and Bacterial Sepsis in Critically Ill Patients. Critical Care 25: 1-12. DOI: https://doi.org/10.1186/s13054-021-03631-4.
https://doi.org/10.1186/s13054-021-03631...
, Ahmadian et al. 2020AHMADIAN E, KHATIBI SMH, SOOFIYANI SR, ABEDIAZAR S, SHOJA MM, ARDALAN M & VAHED SZ. 2020. Covid-19 and Kidney Injury: Pathophysiology and Molecular Mechanisms. Rev Med Virol 31(3): e2176. Accessed July 1, 2020. https://doi.org/10.1002/rmv.2176.
https://doi.org/10.1002/rmv.2176...
).

During cytokine storms, IL-6 plays an important role in the establishment of lesions due to its ability to induce the recruitment of other pro-inflammatory cytokines and chemokines (Machado et al. 2004MACHADO PRL, ARAÚJO MIAS, CARVALHO L & CARVALHO EM. 2004. Mecanismos de Resposta Imune às Infecções. An Bras Dermatol 79(6): 647-662., Brandão et al. 2020BRANDÃO SCS, GODOI ETAM, RAMOS JOX, MELO LMMP & SARINHO ESC. 2020. Severe COVID-19: Understanding the Role of Immunity, Endothelium, and Coagulation in Clinical Practice. J Vasc Bras 19: e20200131.). Many studies have demonstrated the presence of elevated inflammatory markers, such as C-reactive protein (CRP), in patients with SARS-CoV-2 (Tarragón et al. 2021TARRAGÓN B, VALDENEBRO M, SERRANO ML, MAROTO A, LLÓPEZ-CARRATALÁ MR, RAMOS A, RUBIO E, HUERTA A, MARQUES M & PORTOLÉS J. 2021. Fracaso Renal Agudo En Pacientes Hospitalizados Por COVID-19. Nefrologia 41(1): 34-40., Taha et al. 2021TAHA M, SANO D, HANOUDI S, ESBER Z, ELAHI M, GABALI A, CHOPRA T, DRAGHICI S & SAMAVATI L. 2021a. Platelets and Renal Failure in the SARS-CoV-2 Syndrome. Platelets 32 (1): 130-137. DOI: https://doi.org/10.1080/09537104.2020.1817361.
https://doi.org/10.1080/09537104.2020.18...
, Cheng et al. 2020aCHENG Y, LUO R, WANG K, ZHANG M, WANG Z, DONG L, LI J, YAO Y, GE S & XU G. 2020a. Kidney Disease Is Associated with in-Hospital Death of Patients with COVID-19. Kidney Int 97(5): 829.). This protein is synthesized by the liver in response to IL-6, which is a biomarker of inflammation widely associated with AKI (Smilowitz et al. 2021SMILOWITZ NR, KUNICHOFF D, GARSHICK M, SHAH B, PILLINGER M, HOCHMAN JS & BERGER JS. 2021. C-Reactive Protein and Clinical Outcomes in Patients with COVID-19. Eur Heart J 42(23): 2270-2779., Tarragón et al. 2021TARRAGÓN B, VALDENEBRO M, SERRANO ML, MAROTO A, LLÓPEZ-CARRATALÁ MR, RAMOS A, RUBIO E, HUERTA A, MARQUES M & PORTOLÉS J. 2021. Fracaso Renal Agudo En Pacientes Hospitalizados Por COVID-19. Nefrologia 41(1): 34-40., Taha et al. 2021TAHA M, SANO D, HANOUDI S, ESBER Z, ELAHI M, GABALI A, CHOPRA T, DRAGHICI S & SAMAVATI L. 2021a. Platelets and Renal Failure in the SARS-CoV-2 Syndrome. Platelets 32 (1): 130-137. DOI: https://doi.org/10.1080/09537104.2020.1817361.
https://doi.org/10.1080/09537104.2020.18...
, Wang et al. 2021WANG J, YANG X, LI Y, HUANG JA, JIANG J & SU N. 2021. Specific Cytokines in the Inflammatory Cytokine Storm of Patients with COVID-19-Associated Acute Respiratory Distress Syndrome and Extrapulmonary Multiple-Organ Dysfunction. Virol J 18: 1-12. DOI: https://doi.org/10.1186/s12985-021-01588-y.
https://doi.org/10.1186/s12985-021-01588...
). The excessive number of inflammatory cells is capable of disrupting the homeostasis of the body, even interfering with the integrity of endothelial cells (Libby & Lüscher 2020LIBBY P & LÜSCHER T. 2020. COVID-19 Is, in the End, an Endothelial Disease. Eur Heart J 41(32): 3038-3044.). Desai et al. (2002)DESAI TR, LEEPER NJ, HYNES KL & GEWERTZ BL. 2002. Interleukin-6 Causes Endothelial Barrier Dysfunction via the Protein Kinase C Pathway. J Surg Res 104(2): 118-123. https://doi.org/10.1006/jsre.2002.6415.
https://doi.org/10.1006/jsre.2002.6415...
demonstrated that the increase in IL-6 concentrations was associated with endothelial barrier dysfunction, followed by an increase in renal vascular permeability. Increased vascular permeability is associated with facilitation in the entry of leukocytes and proteins, with consequent formation of edema in the renal interstitium, to increase the distance by which oxygen should diffuse and reach tubular tissue (Kinsey et al. 2008KINSEY GR, LI L & OKUSA MD. 2008. Inflammation in Acute Kidney Injury. Nephron Exp Nephrol 109(4): e102-e107.). In addition, cytokines and viral particles activate endothelial cells, which positively regulate the expression of adhesion molecules and monocyte chemoattractants. This leads to the recruitment of activated monocytes to the endothelial cells, as well as increased adherence of leukocytes and leukocyte-endothelium interactions, which allows the passage of leukocytes toward the renal interstitium. Leukocytes coming out of peritubular capillaries are close to tubular epithelial cells and can directly induce the injury of these cells (Ostermann & Liu 2017OSTERMANN M & LIU K. 2017. Pathophysiology of AKI. Best Pract Res Clin Anaesthesiol 31(3): 305-314.) (Figure 3).

Hemodynamic instability

In addition to the tissue lesions themselves, cytokine storms can cause hemodynamic instability, with reduced renal perfusion and consequent damage associated with hypoxia and tissue ischemia (Chong & Saha 2021, Chueh et al. 2020CHUEH TI, ZHENG CM, HOU YC & LU KC. 2020. Novel Evidence of Acute Kidney Injury in COVID-19. J Clin Med Res 9(11): 3547. DOI: https://doi.org/10.3390/jcm9113547.
https://doi.org/10.3390/jcm9113547...
). Sites with low oxygen concentrations may coexist with oxygenated regions of preserved tissue. Interactions between these areas are associated with increased generation of reactive oxygen species (ROS), which may further impair arteriolar blood flow, inducing damage to endothelial cells (Ostermann & Liu 2017OSTERMANN M & LIU K. 2017. Pathophysiology of AKI. Best Pract Res Clin Anaesthesiol 31(3): 305-314., Legrand et al. 2008LEGRAND M, MIK EG, JOHANNES T, PAYEN D & INCE C. 2008. Renal Hypoxia and Dysoxia After Reperfusion of the Ischemic Kidney. Mol Med 14(7): 502-516.). It is also possible that hyperactivation of the classic axis of RAAS contributes to the depletion of renal perfusion. As illustrated in Figure 2, the downregulation of ACE-2 from the entry of SARS-CoV-2 in the intracellular medium favors the accumulation of circulating Ang II, which has vasoconstrictor, profibrotic, and pro-inflammatory effects, further reducing renal blood flow and enabling ischemia (Ames et al. 2019AMES MK, ATKINS CE & PITT B. 2019. The Renin-Angiotensin-Aldosterone System and Its Suppression. J Vet Intern Med 33(2): 363-382., Mizuiri & Ohashi 2015MIZUIRI S & OHASHI Y. 2015. ACE and ACE2 in Kidney Disease. World J Nephrol 4(1): 74-82.).

In a single-center study in the United States (USA), more than 60% of AKI cases were attributed to acute tubular injury due to ischemia or toxicity (Mohamed et al. 2020MOHAMED MMB, LUKITSCH I, TORRES-ORTIZ AE, WALKER JB, VARGHESE V, HERNANDEZ-ARROYO CF, ALQUDSI M, LEDOUX JR & VELEZ JCQ. 2020. Acute Kidney Injury Associated with Coronavirus Disease 2019 in Urban New Orleans. Kidney360 1(7): 614. DOI: https://doi.org/10.34067/kid.0002652020.
https://doi.org/10.34067/kid.0002652020...
). In this sense, it is possible that the AKI observed in patients with COVID-19 may have one of its etiologies based on impaired perfusion of the kidneys. Thus, renal tissue biopsies revealed a diffuse lesion of the proximal tubule with ATN as the most common pathological alteration observed in patients with COVID-19 who developed AKI (Su et al. 2020SU H ET AL. 2020. Renal Histopathological Analysis of 26 Postmortem Findings of Patients with COVID-19 in China. Kidney Int 98(1): 219-227., Chueh et al. 2020CHUEH TI, ZHENG CM, HOU YC & LU KC. 2020. Novel Evidence of Acute Kidney Injury in COVID-19. J Clin Med Res 9(11): 3547. DOI: https://doi.org/10.3390/jcm9113547.
https://doi.org/10.3390/jcm9113547...
).

Dysregulation of complement

During COVID-19 infection, in addition to the inflammatory response mediated by cytokines and chemokines, there is also the participation of an integral component of the innate immune response, the complement system. This system consists of 3 main routes: classical route, alternative route, and lectin pathway. All these pathways converge in the cleavage of C3 in C3a (inflammation) and C3b (phagocytosis and opsonization). The C3b recruits the C5, cleaving it in C5a, which is a potent anaphylatoxin, as well as in C5b that is responsible for the formation of the membrane attack complex (MAC)/C5b-9 (Sim & Tsiftsoglou 2004SIM RB & TSIFTSOGLOU SA. 2004. Proteases of the Complement System. Biochem Soc Trans 32(1): 21-27. DOI: https://doi.org/10.1042/bst0320021.
https://doi.org/10.1042/bst0320021...
). The activation of this system at the physiological level may favor the elimination of pathogens. However, when hyperactivated results in acute and chronic inflammation, tissue injury, and coagulation activation (Noris et al. 2020NORIS M, BENIGNI A & REMUZZI G. 2020. The Case of Complement Activation in COVID-19 Multiorgan Impact. Kidney Int 98(2): 314-322., Wang et al. 2015WANG R, XIAO H, GUO R, LI Y & SHEN B. 2015. The Role of C5a in Acute Lung Injury Induced by Highly Pathogenic Viral Infections. Emerg Microbes & Infect 4(5): 1-7.). High levels of C5a have been produced as a result of excessive complement activation and are involved in the recruitment of inflammatory cells such as neutrophils, eosinophils, monocytes, and T lymphocytes, activation of phagocytic cells, the release of enzymes based on granules, and generation of pro-oxidant agents (Guo & Ward 2005GUO RF & WARD PA. 2005. Role of C5a in inflammatory responses. Annu Rev Immunol 23: 821-852. DOI: https://doi.org/10.1146/annurev.immunol.23.021704.115835.
https://doi.org/10.1146/annurev.immunol....
).

Currently, experimental models are lacking that explain the mechanisms by which the complement system is associated with the damage caused in the course of SARS-CoV-2. For this, some studies suggest mechanisms of injury based on previous analyses in the context of SARS and MERS infection. In an experimental study with C3 deficient mice infected with SARS-CoV, a less severe form of the disease was noted, where this group presented lower weight loss, respiratory dysfunction, and cytokine and chemokine levels compared to the group of wild mice (Gralinski et al. 2018GRALINSKI LE, SHEAHAN TP, MORRISON TE, MENACHERY VD, JENSEN K, LEIST SR, WHITMORE A, HEISE MT & BARIC RS. 2018. Complement Activation Contributes to Severe Acute Respiratory Syndrome Coronavirus Pathogenesis. mBio 9(5): e01753-18. DOI: https://doi.org/10.1128/mbio.01753-18.
https://doi.org/10.1128/mbio.01753-18...
). Jiang et al. (2018)JIANG Y ET AL. 2018. Blockade of the C5a–C5aR Axis Alleviates Lung Damage in hDPP4-Transgenic Mice Infected with MERS-CoV. Emerg Microbes & Infect 7(1): 1-12. DOI: https://doi.org/10.1038/s41426-018-0063-8.
https://doi.org/10.1038/s41426-018-0063-...
evaluated complement cell levels in MERS-CoV-infected mice, the results showed that circulating C5a concentrations increased, as well as the expression of their receptor (C5aR), associated with strong deposition of C5b-9 in bronchiolar epithelial cells, pneumocytes, and infiltrating leukocytes. Although the reported studies are related to SARS and MERS, as well as the observed elements are restricted to the respiratory system, these data together may provide additional information about COVID-19, even suggesting that the complement may affect other organs, including the kidneys. In line with this possibility, Diao et al. (2020)DIAO B ET AL. 2020. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) medRxiv. 2020: 2020.03. 04.20031120 noticed through strong immunohistochemistry C5b-9 deposition in renal tissues of patients with AKI, when compared to patients without kidney injury, suggesting that SARS-CoV-2 infection induces NTA through the deposition of C5b-9.

Rhabdomyolysis

Rhabdomyolysis is a clinical syndrome in which skeletal muscle damage arising from direct traumatic injury, drugs, toxins, muscle ischemia, electrolyte, metabolic disorders, and infections results in the release of muscle contents into the circulatory system. This released intracellular content includes creatine kinase (CK), aldolase, lactate dehydrogenase (LDH), and myoglobin, a potentially nephrotoxic protein, leading to renal dysfunction (Souza et al. 2020SOUZA SI ET AL. 2021. Acute Kidney Injury Induced by Glycerol Is Worsened by Orchiectomy and Attenuated by Testosterone Replacement. Steroids 165(January): 108755., Torres et al. 2015TORRES PA, HELMSTETTER JA, KAYE AM & KAYE AD. 2015. Rhabdomyolysis: Pathogenesis, Diagnosis, and Treatment. Ochsner J 15(1): 58-69.). Viral infections can also cause muscle disorders, but influenza is the most common virus associated with rhabdomyolysis (Singh & Scheid 1996SINGH U & SCHELD WM. 1996. Infectious etiologies of rhabdomyolysis: three case reports and review. Clin Infect Dis 22(4): 642-649. DOI: 10.1093/clinids/22.4.642. PMID: 8729203., Suwanwongse & Shabarek 2020SUWANWONGSE K & SHABAREK M. 2020. Rhabdomyolysis as a Presentation of 2019 Novel Coronavirus Disease. Cureus 12(4): e7561. DOI: https://doi.org/10.7759/cureus.7561.
https://doi.org/10.7759/cureus.7561...
). Recently, the presence of rhabdomyolysis has been reported in patients with COVID-19. Rhabdomyolysis may be the initial presentation of SARS-CoV-2 or occur at any time during the disease course (Suwanwongse & Shabarek 2020SUWANWONGSE K & SHABAREK M. 2020. Rhabdomyolysis as a Presentation of 2019 Novel Coronavirus Disease. Cureus 12(4): e7561. DOI: https://doi.org/10.7759/cureus.7561.
https://doi.org/10.7759/cureus.7561...
). A single-center retrospective observational study compared laboratory alterations found in patients infected with COVID-19 with and without AKI. Compared to patients without AKI, AKI patients had higher CK and LDH counts (Guanhua et al. 2021GUANHUA X ET AL. 2021. Acute Kidney Injury in Patients Hospitalized with COVID-19 in Wuhan, China: A Single-Center Retrospective Observational Study. J South Med Univ 41(2): 157.). Although this study did not evaluate the appearance of rhabdomyolysis preceding AKI, the alterations found are consistent with those induced by rhabdomyolysis.

Some hypotheses have been postulated to report the pathogenesis of rhabdomyolysis during a viral infection. First, direct invasion of the virus can lead to rhabdomyolysis. Secondly, the cytokine storm can cause muscle damage. Finally, circulating toxins can directly destroy muscle cell membranes (Suwanwongse & Shabarek 2020SUWANWONGSE K & SHABAREK M. 2020. Rhabdomyolysis as a Presentation of 2019 Novel Coronavirus Disease. Cureus 12(4): e7561. DOI: https://doi.org/10.7759/cureus.7561.
https://doi.org/10.7759/cureus.7561...
). However, the exact mechanism of COVID-19-induced rhabdomyolysis has not yet been proposed, but direct invasion and hypercytokinemia during SARS-CoV-2 resemble the mechanisms reported for other viral infections.

Endothelial dysfunction and hypercoagulation

The inflammatory response generated by SARS-CoV-2 is strongly related to the increase in coagulatory processes. This increased state of coagulation is almost always mediated by the tissue factor (TF) pathway. Usually, TF is not expressed in monocytes and endothelial cells. However, due to endothelial injury or specific stimuli from endotoxins and cytokines, these cells begin to express the TF (Merad & Martin 2020MERAD M & MARTIN JC. 2020. Pathological Inflammation in Patients with COVID-19: A Key Role for Monocytes and Macrophages. Nat Rev Immunol 20(6): 355-362.). The TF forms a complex with factor VII, stimulating the conversion of factor X into Xa. The latter factor plays a crucial role in the coagulation cascade, converting prothrombin into thrombin along with its cofactor Va. Large amounts of thrombin are formed, causing additional platelet activation, intensified fibrin formation from hepatic fibrinogen, and thrombus deposition at the injured site (Witkowski et al. 2016WITKOWSKI M, LANDMESSER U & RAUCH U. 2016. Tissue Factor as a Link between Inflammation and Coagulation. Trends Cardiovasc Med 26(4): 297-303. DOI: https://doi.org/10.1016/j.tcm.2015.12.001.
https://doi.org/10.1016/j.tcm.2015.12.00...
). Tissue factor inhibitor (TFPI), the main anticoagulant pathway, is almost always impaired in an inflammatory process, further accentuating the procoagulant state in the body (Merad & Martin 2020MERAD M & MARTIN JC. 2020. Pathological Inflammation in Patients with COVID-19: A Key Role for Monocytes and Macrophages. Nat Rev Immunol 20(6): 355-362.). Thrombosis plays a central role in COVID-19-related AKI since SARS-CoV-2 infection can trigger the activation of the coagulation cascade leading to renal vascular injury, with the development of ischemic glomeruli and fibrinoid necrosis (Cheng et al. 2020bCHENG Y, LUO R, WANG K, ZHANG M, WANG Z, DONG L, LI J, YAO Y, GE S & XU G. 2020b. Kidney Impairment Is Associated with in-Hospital Death of COVID-19 Patients. DOI: https://doi.org/10.1101/2020.02.18.20023242.
https://doi.org/10.1101/2020.02.18.20023...
). COVID-19 may predispose to venous and arterial thromboembolic disease due to excessive inflammation, hypoxia, immobilization, and diffuse intravascular coagulation (Klok et al. 2020KLOK FA ET AL. 2020. Incidence of Thrombotic Complications in Critically Ill ICU Patients with COVID-19. Thromb Res 191: 145-147.).

Abnormalities involved in coagulation are associated with a poor prognosis and may represent the main cause of organ failure and death in patients with COVID-19 (Merad & Martin 2020MERAD M & MARTIN JC. 2020. Pathological Inflammation in Patients with COVID-19: A Key Role for Monocytes and Macrophages. Nat Rev Immunol 20(6): 355-362.). Tang et al. (2020)TANG N, LI D, WANG X & SUN Z. 2020. Abnormal Coagulation Parameters Are Associated with Poor Prognosis in Patients with Novel Coronavirus Pneumonia. J Thromb Haemost 18(4): 844-847. revealed that 21 of the 183 patients with COVID-19 died, and this group had higher levels of D dimer and fibrin degradation product (DDP), longer prothrombin time, and part-time thromboplastin activated compared to survivors on admission. In a study with 701 patients, 81.6% had high levels of erythrocyte sedimentation (Cheng et al. 2020aCHENG Y, LUO R, WANG K, ZHANG M, WANG Z, DONG L, LI J, YAO Y, GE S & XU G. 2020a. Kidney Disease Is Associated with in-Hospital Death of Patients with COVID-19. Kidney Int 97(5): 829.). Nevertheless, coagulation pathway abnormalities, including prolonged activation in Partial Thromboplastin Time and increased D-dimer, were more common in patients with elevated creatinine (Cheng et al. 2020aCHENG Y, LUO R, WANG K, ZHANG M, WANG Z, DONG L, LI J, YAO Y, GE S & XU G. 2020a. Kidney Disease Is Associated with in-Hospital Death of Patients with COVID-19. Kidney Int 97(5): 829.). Postmortem findings by Su et al. (2020)SU H ET AL. 2020. Renal Histopathological Analysis of 26 Postmortem Findings of Patients with COVID-19 in China. Kidney Int 98(1): 219-227. in patients with SARS-CoV-2 revealed a diffuse lesion of the proximal tubule with loss of brush edge, vacuolar degeneration, and frank necrosis on light microscopy, as well as aggregates of erythrocytes obstructing the lumen of the capillaries without the presence of platelet material (Figure 3). These findings favor ischemic injury and hypoxia of the kidneys by hypoperfusion and show the recurrence of coagulopathies in patients with COVID-19, as well as its important contribution to the onset of AKI (Cheng et al. 2020, Su et al. 2020a).

Finally, both the damage caused by infiltration of the virus into the renal parenchyma and the damage caused by indirect mechanisms are related to the findings of proteinuria and other biomarkers of kidney injury in infected patients, including elevation of serum creatinine and blood urea nitrogen (Rapkiewicz et al. 2020RAPKIEWICZ AV ET AL. 2020. Megakaryocytes and Platelet-Fibrin Thrombi Characterize Multi-Organ Thrombosis at Autopsy in COVID-19: A Case Series. eClinicalMedicine 24(July): 100434., Cao et al. 2020CAO M ET AL. 2020. Clinical Features of Patients Infected with the 2019 Novel Coronavirus (COVID-19) in Shanghai, China. medRxiv : The Preprint Server for Health Sciences, March. https://doi.org/10.1101/2020.03.04.20030395.
https://doi.org/10.1101/2020.03.04.20030...
). In addition, tubular damage itself may be associated with a decrease in glomerular filtration rate (GFR), despite the lack of damage to the glomeruli itself. In the case of damage or dysfunction of tubular cells, sodium reabsorption in the proximal tubule is impaired by increasing the concentrations of sodium chloride arriving in the dense macula. This triggers tubuloglomerular feedback and leads to preglomerular vasoconstriction of the afferent arteriole, resulting in a decline in GFR (Ostermann & Liu 2017OSTERMANN M & LIU K. 2017. Pathophysiology of AKI. Best Pract Res Clin Anaesthesiol 31(3): 305-314.).

CONCLUSION

Acute kidney injury is a common complication in patients with COVID-19, with a high impact on morbidity and mortality, primarily characterized by acute tubular necrosis and renal dysfunctions. To date, the available studies suggest the involvement of a series of pathophysiological pathways in the development of lesions, which can be activated by the direct action of the virus in the cells, leading to hyperactivation of the classic axis of RAAS and repercussions in KKS, and by indirect mechanisms resulting from the repercussions of systemic viral infection that trigger an exaggerated inflammatory response, hemodynamic changes, tissue hypoxia, generation of ROS, endothelial damage and coagulopathies.

As perspectives, the elucidation of molecular pathways with the identification of key molecules involved in tissue damage is extremely important for the prevention and treatment of AKI, since it can contribute to the advancement of other research intended at the development of new drugs aimed at blocking the entry of the virus into the cell, as well as in the control of immune responses.

ACKNOWLEDGMENTS

This work was supported by the Institutional Program of Scientific Initiation Scholarship (PIBIC) from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação de Amparo à Pesquisa do Estado da Bahia (FAPESB).

REFERENCES

  • ADAPA S, CENNA A, BALLA M, MERUGU GP, KODURI NM, DAGGUBATI SR, GAYAM V, NARAMALA S & KONALA VM. 2020. COVID-19 Pandemic Causing Acute Kidney Injury and Impact on Patients With Chronic Kidney Disease and Renal Transplantation. J Clin Med Res 12(6): 352-361.
  • AHMADIAN E, KHATIBI SMH, SOOFIYANI SR, ABEDIAZAR S, SHOJA MM, ARDALAN M & VAHED SZ. 2020. Covid-19 and Kidney Injury: Pathophysiology and Molecular Mechanisms. Rev Med Virol 31(3): e2176. Accessed July 1, 2020. https://doi.org/10.1002/rmv.2176.
    » https://doi.org/10.1002/rmv.2176
  • ALMEIDA DC ET AL. 2021. Acute Kidney Injury: Incidence, Risk Factors, and Outcomes in Severe COVID-19 Patients. PLoS ONE 16(5): e0251048.
  • AMES MK, ATKINS CE & PITT B. 2019. The Renin-Angiotensin-Aldosterone System and Its Suppression. J Vet Intern Med 33(2): 363-382.
  • BATLLE D, SOLER MJ, SPARKS MA, HIREMATH S, SOUTH AM, WELLING PA & SWAMINATHAN S. 2020. Acute Kidney Injury in COVID-19: Emerging Evidence of a Distinct Pathophysiology. J Am Soc Nephrol 31(7): 1380-1383. https://doi.org/10.1681/ASN.2020040419.
    » https://doi.org/10.1681/ASN.2020040419
  • BRADLEY BT ET AL. 2020. Histopathology and Ultrastructural Findings of Fatal COVID-19 Infections in Washington State: A Case Series. Lancet 396(10247): 320-332.
  • BRANDÃO SCS, GODOI ETAM, RAMOS JOX, MELO LMMP & SARINHO ESC. 2020. Severe COVID-19: Understanding the Role of Immunity, Endothelium, and Coagulation in Clinical Practice. J Vasc Bras 19: e20200131.
  • CACERES PS ET AL. 2021. High SARS-CoV-2 Viral Load in Urine Sediment Correlates with Acute Kidney Injury and Poor COVID-19 Outcome. J Am Soc Nephrol 32(10): 2517-2528.
  • CAO M ET AL. 2020. Clinical Features of Patients Infected with the 2019 Novel Coronavirus (COVID-19) in Shanghai, China. medRxiv : The Preprint Server for Health Sciences, March. https://doi.org/10.1101/2020.03.04.20030395
    » https://doi.org/10.1101/2020.03.04.20030395
  • CHANNAPPANAVAR R ET AL. 2019. IFN-I Response Timing Relative to Virus Replication Determines MERS Coronavirus Infection Outcomes. J Clin Investig 129(9): 3625.
  • CHANNAPPANAVAR R, FEHR AR, VIJAY R, MACK M, ZHAO J, MEYERHOLZ DK & PERLMAN S. 2016. Dysregulated Type I Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice. Cell Host Microbe 19(2): 181-193.
  • CHEN G ET AL. 2020. Clinical and Immunological Features of Severe and Moderate Coronavirus Disease 2019. J Clin Investig 130(5): 2620-2629.
  • CHENG Y, LUO R, WANG K, ZHANG M, WANG Z, DONG L, LI J, YAO Y, GE S & XU G. 2020a. Kidney Disease Is Associated with in-Hospital Death of Patients with COVID-19. Kidney Int 97(5): 829.
  • CHENG Y, LUO R, WANG K, ZHANG M, WANG Z, DONG L, LI J, YAO Y, GE S & XU G. 2020b. Kidney Impairment Is Associated with in-Hospital Death of COVID-19 Patients. DOI: https://doi.org/10.1101/2020.02.18.20023242
    » https://doi.org/10.1101/2020.02.18.20023242
  • CHU KH ET AL. 2005. Acute Renal Impairment in Coronavirus-Associated Severe Acute Respiratory Syndrome. Kidney Int 67(2): 698-705.
  • CHUEH TI, ZHENG CM, HOU YC & LU KC. 2020. Novel Evidence of Acute Kidney Injury in COVID-19. J Clin Med Res 9(11): 3547. DOI: https://doi.org/10.3390/jcm9113547.
    » https://doi.org/10.3390/jcm9113547
  • CUMMINGS MJ ET AL. 2020. Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 in New York City: a prospective cohort study. Lancet 395(10239): 1763-1770. DOI: 10.1016/S0140-6736(20)31189-2.
  • DESAI TR, LEEPER NJ, HYNES KL & GEWERTZ BL. 2002. Interleukin-6 Causes Endothelial Barrier Dysfunction via the Protein Kinase C Pathway. J Surg Res 104(2): 118-123. https://doi.org/10.1006/jsre.2002.6415.
    » https://doi.org/10.1006/jsre.2002.6415
  • DIAO B ET AL. 2020. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) medRxiv. 2020: 2020.03. 04.20031120
  • DIAO B ET AL. 2021. Human Kidney Is a Target for Novel Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Nat Comm 12(1): 2506.
  • DONOGHUE M ET AL. 2000. A Novel Angiotensin-Converting Enzyme-Related Carboxypeptidase (ACE2) Converts Angiotensin I to Angiotensin 1-9. Circ Res 87(5): E1-E9.
  • EL-SAYED EE, ALLAYEH AK, SALEM AA, OMAR SM, ZAGHLOL SM, ABD-ELMAGUID HM, ABDUL-GHAFFAR MM & ELSHARKAWY MM. 2021. Incidence of Acute Kidney Injury among COVID-19 Patients in Egypt. Ren Replace Ther 7(1): 1-7.
  • FERREIRA AJ & SANTOS RAS. 2005. Cardiovascular Actions of Angiotensin-(1-7). Rev Bras Pesqui Med Biol 38(4): 499-507.
  • GIRARDI ACC, DEGRAY BC, NAGY T, BIEMESDERFER D & ARONSON PS. 2001. Association of Na+-H+ Exchanger Isoform NHE3 and Dipeptidyl Peptidase IV in the Renal Proximal Tubule. J Biol Chem 276(49): 46671-46677.
  • GOLMAI P, LARSEN CP, DEVITA MV, WAHL JS, WEINS A, RENNKE HG, BIJOL V & ROSENSTOCK JL. 2020. Histopathologic and Ultrastructural Findings in Postmortem Kidney Biopsy Material in 12 Patients with AKI and COVID-19. J Am Soc Nephrol 31(9): 1944-1947.
  • GÓMEZ-ESCOBAR LG ET AL. 2021. Cytokine Signatures of End Organ Injury in COVID-19. Sci Rep 11(1): 1-15.
  • GRALINSKI LE, SHEAHAN TP, MORRISON TE, MENACHERY VD, JENSEN K, LEIST SR, WHITMORE A, HEISE MT & BARIC RS. 2018. Complement Activation Contributes to Severe Acute Respiratory Syndrome Coronavirus Pathogenesis. mBio 9(5): e01753-18. DOI: https://doi.org/10.1128/mbio.01753-18.
    » https://doi.org/10.1128/mbio.01753-18
  • GUAN WJ ET AL. 2020. Clinical Characteristics of Coronavirus Disease 2019 in China. N Engl J Med 382(18): 1708-1720. DOI: https://doi.org/10.1056/NEJMoa2002032.
    » https://doi.org/10.1056/NEJMoa2002032
  • GUANHUA X ET AL. 2021. Acute Kidney Injury in Patients Hospitalized with COVID-19 in Wuhan, China: A Single-Center Retrospective Observational Study. J South Med Univ 41(2): 157.
  • GUO RF & WARD PA. 2005. Role of C5a in inflammatory responses. Annu Rev Immunol 23: 821-852. DOI: https://doi.org/10.1146/annurev.immunol.23.021704.115835.
    » https://doi.org/10.1146/annurev.immunol.23.021704.115835
  • HAMMING IWT, BULTHUIS MLC, LELY AT, NAVIS GJ & VAN GOOR H. 2004. Tissue Distribution of ACE2 Protein, the Functional Receptor for SARS Coronavirus. A First Step in Understanding SARS Pathogenesis. J Pathol 203(2): 631-637.
  • HOFFMANN M ET AL. 2020 SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Cell 181(2): 271-280.
  • HUANG C ET AL. 2020. Clinical Features of Patients Infected with 2019 Novel Coronavirus in Wuhan, China. Lancet 395(10223): 497-506.
  • JIANG F, YANG J, ZHANG Y, DONG M, WANG S, ZHANG Q, LIU FF, ZHANG K & ZHANG C. 2014. Angiotensin-Converting Enzyme 2 and Angiotensin 1-7: Novel Therapeutic Targets. Nat Rev Cardiol 11(7): 413-426.
  • JIANG Y ET AL. 2018. Blockade of the C5a–C5aR Axis Alleviates Lung Damage in hDPP4-Transgenic Mice Infected with MERS-CoV. Emerg Microbes & Infect 7(1): 1-12. DOI: https://doi.org/10.1038/s41426-018-0063-8.
    » https://doi.org/10.1038/s41426-018-0063-8
  • KATO N, YUZAWA Y, KOSUGI T, HOBO A, SATO W, MIWA Y, SAKAMOTO K, MATSUO S & KADOMATSU K. 2009. The E-Selectin Ligand Basigin/CD147 Is Responsible for Neutrophil Recruitment in Renal Ischemia/Reperfusion. J Am Soc Nephrol 20(7): 1565-1576.
  • KDIGO - KIDNEY DISEASE: IMPROVING GLOBAL OUTCOMES. 2012. KDIGO Clinical Practice Guideline for Acute Kidney Injury. Kidney Int Suppl 2: 1-138.
  • KINSEY GR, LI L & OKUSA MD. 2008. Inflammation in Acute Kidney Injury. Nephron Exp Nephrol 109(4): e102-e107.
  • KLOK FA ET AL. 2020. Incidence of Thrombotic Complications in Critically Ill ICU Patients with COVID-19. Thromb Res 191: 145-147.
  • KOSUGI T, MAEDA K, SATO W, MARUYAMA S & KADOMATSU K. 2015. CD147 (EMMPRIN/Basigin) in Kidney Diseases: From an Inflammation and Immune System Viewpoint. Nephrol Dial Transplant 30(7): 1097-1103.
  • KUDOSE S ET AL. 2020. Kidney Biopsy Findings in Patients with COVID-19. J Am Soc Nephrol 31(9): 1959-1968.
  • LEGRAND M, MIK EG, JOHANNES T, PAYEN D & INCE C. 2008. Renal Hypoxia and Dysoxia After Reperfusion of the Ischemic Kidney. Mol Med 14(7): 502-516.
  • LI G ET AL. 2020a. Coronavirus Infections and Immune Responses. J Med Virol 92 (4): 424-432.
  • LI T, QUAN H, ZHANG H, LIN L, OU Q & CHEN K. 2020b. Silencing Cyclophilin A Improves Insulin Secretion, Reduces Cell Apoptosis, and Alleviates Inflammation as Well as Oxidant Stress in High Glucose-Induced Pancreatic β-Cells via MAPK/NF-Kb Signaling Pathway. Bioengineered 11(1): 1047-1057. DOI: https://doi.org/10.1080/21655979.2020.1823729.
  • LI W ET AL. 2003. Angiotensin-Converting Enzyme 2 Is a Functional Receptor for the SARS Coronavirus. Nature 426(6965): 450-454. DOI: https://doi.org/10.1038/nature02145.
    » https://doi.org/10.1038/nature02145
  • LI Y, ZHOU W, YANG L & YOU R. 2020c. Physiological and Pathological Regulation of ACE2, the SARS-CoV-2 Receptor. Pharmacol Res 157: 104833.
  • LI Z ET AL. 2020d. Caution on Kidney Dysfunctions of COVID-19 Patients. medRxiv 2020.02.08.20021212. DOI: https://doi.org/10.1101/2020.02.08.20021212
    » https://doi.org/10.1101/2020.02.08.20021212
  • LIAO Y, LUO D, PENG K & ZENG Y. 2021. Cyclophilin A: A Key Player for Etiological Agent Infection. Appl Microbiol Biotechnol 105(4): 1365-1377. DOI: https://doi.org/10.1007/s00253-021-11115-2.
    » https://doi.org/10.1007/s00253-021-11115-2
  • LIBBY P & LÜSCHER T. 2020. COVID-19 Is, in the End, an Endothelial Disease. Eur Heart J 41(32): 3038-3044.
  • LIONEL BI & DONLIN LT. 2014. Regulation of Type I Interferon Responses. Nat Rev Immunol 14(1): 36-49.
  • LIU X, ZHAO Z & LIU W. 2013. Insights into the Roles of Cyclophilin A during Influenza Virus Infection. Viruses 5(1): 182-191. DOI: https://doi.org/10.3390/v5010182.
    » https://doi.org/10.3390/v5010182
  • LUCAS JM, TRUE L, HAWLEY S, MATSUMURA M, MORRISSEY C, VESSELLA R & NELSON PS. 2008. The Androgen-Regulated Type II Serine Protease TMPRSS2 Is Differentially Expressed and Mislocalized in Prostate Adenocarcinoma. J Pathol 215(2): 118-125. https://doi.org/10.1002/path.2330.
    » https://doi.org/10.1002/path.2330
  • MACHADO PRL, ARAÚJO MIAS, CARVALHO L & CARVALHO EM. 2004. Mecanismos de Resposta Imune às Infecções. An Bras Dermatol 79(6): 647-662.
  • MAHMUDPOUR M, ROOZBEH J, KESHAVARZ M, FARROKHI S & NABIPOUR I. 2020. COVID-19 Cytokine Storm: The Anger of Inflammation. Cytokine 133: 155151. https://doi.org/10.1016/j.cyto.2020.155151.
    » https://doi.org/10.1016/j.cyto.2020.155151
  • MERAD M & MARTIN JC. 2020. Pathological Inflammation in Patients with COVID-19: A Key Role for Monocytes and Macrophages. Nat Rev Immunol 20(6): 355-362.
  • MIZUIRI S & OHASHI Y. 2015. ACE and ACE2 in Kidney Disease. World J Nephrol 4(1): 74-82.
  • MOHAMED MMB, LUKITSCH I, TORRES-ORTIZ AE, WALKER JB, VARGHESE V, HERNANDEZ-ARROYO CF, ALQUDSI M, LEDOUX JR & VELEZ JCQ. 2020. Acute Kidney Injury Associated with Coronavirus Disease 2019 in Urban New Orleans. Kidney360 1(7): 614. DOI: https://doi.org/10.34067/kid.0002652020.
    » https://doi.org/10.34067/kid.0002652020
  • NG JH, BIJOL V, SPARKS MA, SISE ME, IZZEDINE H & JHAVERI KD. 2020. Pathophysiology and Pathology of Acute Kidney Injury in Patients With COVID-19. Adv Chronic Kidney Dis 27(5): 365-376.
  • NICOLAU LAD, MAGALHÃES PJC & VALE ML. 2020. What Would Sérgio Ferreira Say to Your Physician in This War against COVID-19: How about Kallikrein/kinin System? Med Hypotheses 143: 109886.
  • NISTALA R & SAVIN V. 2017. Diabetes, Hypertension, and Chronic Kidney Disease Progression: Role of DPP4. Am J Physiol Renal Physiol 312(4): F661-F670. DOI: https://doi.org/10.1152/ajprenal.00316.2016.
    » https://doi.org/10.1152/ajprenal.00316.2016
  • NORIS M, BENIGNI A & REMUZZI G. 2020. The Case of Complement Activation in COVID-19 Multiorgan Impact. Kidney Int 98(2): 314-322.
  • OLIVEIRA LCG, CRUZ NAN, RICELLI B, TEDESCO-SILVA H, MEDINA-PESTANA JO & CASARINI DE. 2021. Interactions amongst Inflammation, Renin-Angiotensin-Aldosterone and Kallikrein-Kinin Systems: Suggestive Approaches for COVID-19 Therapy. J Venom Anim Toxins Incl Trop Dis 27(December). DOI: https://doi.org/10.1590/1678-9199-JVATITD-2020-0181
    » https://doi.org/10.1590/1678-9199-JVATITD-2020-0181
  • OSTERMANN M & LIU K. 2017. Pathophysiology of AKI. Best Pract Res Clin Anaesthesiol 31(3): 305-314.
  • PAN XW, XU D, ZHANG H, ZHOU W, WANG LH & CUI XG. 2020. 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 46(6): 1114-1116.
  • PERICO L, BENIGNI A & REMUZZI G. 2020. Should COVID-19 Concern Nephrologists? Why and to What Extent? The Emerging Impasse of Angiotensin Blockade. Nephron 144(5): 213-221.
  • PESQUERO JB & BADER M. 1998. Molecular Biology of the Kallikrein-Kinin System: From Structure to Function. Rev Med Pesqui Med Biol 31(9): 1197-1203.
  • POVLSEN AL, GRIMM D, WEHLAND M, INFANGER M & KRÜGER M. 2020. The Vasoactive Mas Receptor in Essential Hypertension. J Clin Med Res 9(1): 267. https://doi.org/10.3390/jcm9010267.
    » https://doi.org/10.3390/jcm9010267
  • QI F, QIAN S, ZHANG S & ZHANG Z. 2020. Single Cell RNA Sequencing of 13 Human Tissues Identify Cell Types and Receptors of Human Coronaviruses. Biochem Biophys Res Commun 526(1): 135-140.
  • QI K ET AL. 2021. Clinical, Laboratory, and Imaging Features of Pediatric COVID-19: A Systematic Review and Meta-Analysis. Medicine 100(15): e25230.
  • QIN C ET AL. 2020. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infec Dis 71(15): 762-768.
  • QU X, WANG C, ZHANG J, QIE G & ZHOU J. 2014. The Roles of CD147 And/or Cyclophilin A in Kidney Diseases. Mediators Inflamm 71(15): 762-768. DOI: https://doi.org/10.1155/2014/728673.
    » https://doi.org/10.1155/2014/728673
  • RAMALHO FS. 2000. A Regeneração Hepática E Os Inibidores Da Enzima Conversora Da Angiotensina. Acta Cir Bras. DOI: https://doi.org/10.1590/s0102-86502000000600004
    » https://doi.org/10.1590/s0102-86502000000600004
  • RAPKIEWICZ AV ET AL. 2020. Megakaryocytes and Platelet-Fibrin Thrombi Characterize Multi-Organ Thrombosis at Autopsy in COVID-19: A Case Series. eClinicalMedicine 24(July): 100434.
  • RONCO C & REIS T. 2020. Kidney Involvement in COVID-19 and Rationale for Extracorporeal Therapies. Nat Rev Nephrol 16(6): 308-310.
  • SANTORIELLO D, KHAIRALLAH P, BOMBACK AS, XU K, KUDOSE S, BATAL I, BARASCH J, RADHAKRISHNAN J, D’AGATI V & MARKOWITZ G. 2020. Postmortem Kidney Pathology Findings in Patients with COVID-19. J Am Soc Nephrol 31(9): 2158-2167.
  • SANTOS RA ET AL. 2003. Angiotensin-(1-7) Is an Endogenous Ligand for the G Protein-Coupled Receptor Mas. Proc Nat Acad Sci USA 100(14): 8258-8263. DOI: https://doi.org/10.1073/pnas.1432869100.
    » https://doi.org/10.1073/pnas.1432869100
  • SHARMA P ET AL. 2020. COVID-19-Associated Kidney Injury: A Case Series of Kidney Biopsy Findings. J Am Soc Nephrol 31(9): 1948-1958.
  • SIM RB & TSIFTSOGLOU SA. 2004. Proteases of the Complement System. Biochem Soc Trans 32(1): 21-27. DOI: https://doi.org/10.1042/bst0320021.
    » https://doi.org/10.1042/bst0320021
  • SINGH U & SCHELD WM. 1996. Infectious etiologies of rhabdomyolysis: three case reports and review. Clin Infect Dis 22(4): 642-649. DOI: 10.1093/clinids/22.4.642. PMID: 8729203.
  • SMILOWITZ NR, KUNICHOFF D, GARSHICK M, SHAH B, PILLINGER M, HOCHMAN JS & BERGER JS. 2021. C-Reactive Protein and Clinical Outcomes in Patients with COVID-19. Eur Heart J 42(23): 2270-2779.
  • SODHI CP, WOHLFORD-LENANE C, YAMAGUCHI Y, PRINDLE T, FULTON WB, WANG S, MCCRAY JR PB, CHAPPELL M, HACKAM DJ & JIA H. 2018. Biomarkers in Lung Diseases: From Pathogenesis to Prediction to New Therapies: Attenuation of Pulmonary ACE2 Activity Impairs Inactivation of Des-Arg9 bradykinin/BKB1R Axis and Facilitates LPS-Induced Neutrophil Infiltration. Am J Physiol Lung Cell Mol Physiol 314(1): L17-L31.
  • SOLEIMANI M. 2020. Acute Kidney Injury in SARS-CoV-2 Infection: Direct Effect of Virus on Kidney Proximal Tubule Cells. Int J Mol Sci 21(9): 3275.
  • SOUZA SI ET AL. 2021. Acute Kidney Injury Induced by Glycerol Is Worsened by Orchiectomy and Attenuated by Testosterone Replacement. Steroids 165(January): 108755.
  • SU H ET AL. 2020. Renal Histopathological Analysis of 26 Postmortem Findings of Patients with COVID-19 in China. Kidney Int 98(1): 219-227.
  • SU H, WAN C, WANG ZD, GAO Y, LI YC, TANG F, ZHU HY, YI LX & ZHANG C. 2021. Expression of CD147 and Cyclophilin A in Kidneys of Patients with COVID-19. Clin J Am Soc Nephrol 16(4): 618-619.
  • SUWANWONGSE K & SHABAREK M. 2020. Rhabdomyolysis as a Presentation of 2019 Novel Coronavirus Disease. Cureus 12(4): e7561. DOI: https://doi.org/10.7759/cureus.7561.
    » https://doi.org/10.7759/cureus.7561
  • SZABOLCS M ET AL. 2021. Identification of Immunohistochemical Reagents for In Situ Protein Expression Analysis of Coronavirus-Associated Changes in Human Tissues. Appl Immunohistochem Mol Morphol 29(1): 5-12.
  • TAHA M, SANO D, HANOUDI S, ESBER Z, ELAHI M, GABALI A, CHOPRA T, DRAGHICI S & SAMAVATI L. 2021a. Platelets and Renal Failure in the SARS-CoV-2 Syndrome. Platelets 32 (1): 130-137. DOI: https://doi.org/10.1080/09537104.2020.1817361.
    » https://doi.org/10.1080/09537104.2020.1817361
  • TANG N, LI D, WANG X & SUN Z. 2020. Abnormal Coagulation Parameters Are Associated with Poor Prognosis in Patients with Novel Coronavirus Pneumonia. J Thromb Haemost 18(4): 844-847.
  • TARRAGÓN B, VALDENEBRO M, SERRANO ML, MAROTO A, LLÓPEZ-CARRATALÁ MR, RAMOS A, RUBIO E, HUERTA A, MARQUES M & PORTOLÉS J. 2021. Fracaso Renal Agudo En Pacientes Hospitalizados Por COVID-19. Nefrologia 41(1): 34-40.
  • TIAN L, LIU W & SUN L. 2020. Role of Cyclophilin A during Coronavirus Replication and the Antiviral Activities of Its Inhibitors. Chin J Biotechnol 36(4): 605-611. DOI: https://doi.org/10.13345/j.cjb.200049.
    » https://doi.org/10.13345/j.cjb.200049
  • TORRES PA, HELMSTETTER JA, KAYE AM & KAYE AD. 2015. Rhabdomyolysis: Pathogenesis, Diagnosis, and Treatment. Ochsner J 15(1): 58-69.
  • TOTURA AL & BARIC RS. 2012. SARS Coronavirus Pathogenesis: Host Innate Immune Responses and Viral Antagonism of Interferon. Curr Opin Virol 2(3): 264.
  • TSUTSUMI Y ET AL. 1999. Angiotensin II Type 2 Receptor Overexpression Activates the Vascular Kinin System and Causes Vasodilation. J Clin Invest 104(7): 925-935. DOI: https://doi.org/10.1172/JCI7886.
    » https://doi.org/10.1172/JCI7886
  • VAARALA MH, PORVARI KS, KELLOKUMPU S, KYLLÖNEN AP & VIHKO PT. 2001. Expression of Transmembrane Serine Protease TMPRSS2 in Mouse and Human Tissues. J Pathol 193(1): 134-140. DOI: https://doi.org/10.1002/1096-9896(2000)9999:9999<::AID-PATH743>3.0.CO;2-T.
    » https://doi.org/10.1002/1096-9896(2000)9999:9999<::AID-PATH743>3.0.CO;2-T
  • VOLBEDA M ET AL. 2021. Comparison of Renal Histopathology and Gene Expression Profiles between Severe COVID-19 and Bacterial Sepsis in Critically Ill Patients. Critical Care 25: 1-12. DOI: https://doi.org/10.1186/s13054-021-03631-4.
    » https://doi.org/10.1186/s13054-021-03631-4
  • WANG J, YANG X, LI Y, HUANG JA, JIANG J & SU N. 2021. Specific Cytokines in the Inflammatory Cytokine Storm of Patients with COVID-19-Associated Acute Respiratory Distress Syndrome and Extrapulmonary Multiple-Organ Dysfunction. Virol J 18: 1-12. DOI: https://doi.org/10.1186/s12985-021-01588-y.
    » https://doi.org/10.1186/s12985-021-01588-y
  • WANG KE ET AL. 2020. SARS-CoV-2 Invades Host Cells via a Novel Route: CD147-Spike Protein. Biorxiv. https://doi.org/10.1101/2020.03.14.988345
    » https://doi.org/10.1101/2020.03.14.988345
  • WANG R, XIAO H, GUO R, LI Y & SHEN B. 2015. The Role of C5a in Acute Lung Injury Induced by Highly Pathogenic Viral Infections. Emerg Microbes & Infect 4(5): 1-7.
  • WERION A ET AL. 2020. SARS-CoV-2 Causes a Specific Dysfunction of the Kidney Proximal Tubule. Kidney Int 98(5): 1296-1307.
  • WITKOWSKI M, LANDMESSER U & RAUCH U. 2016. Tissue Factor as a Link between Inflammation and Coagulation. Trends Cardiovasc Med 26(4): 297-303. DOI: https://doi.org/10.1016/j.tcm.2015.12.001.
    » https://doi.org/10.1016/j.tcm.2015.12.001
  • WOON HC & SAHA BK. 2021. Relationship Between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and the Etiology of Acute Kidney Injury (AKI). Am J Med Sci 361 (3): 287-296.
  • WU H ET AL. 2020. AKI and Collapsing Glomerulopathy Associated with COVID-19 and High-Risk Genotype. J Am Soc Nephrol 31(8): 1688-1695.
  • XU Z, LI W, HAN J, ZOU C, HUANG W, YU W, SHAN X, LUM H, LI X & LIANG G. 2017. Angiotensin II Induces Kidney Inflammatory Injury and Fibrosis through Binding to Myeloid Differentiation Protein-2 (MD2). Sci Rep 7(1): 1-11. https://doi.org/10.1038/srep44911.
    » https://doi.org/10.1038/srep44911
  • YE Q, WANG B & MAO J. 2020. The Pathogenesis and Treatment of the “Cytokine Storm” in COVID-19. J Infect 80(6): 607-613.
  • YIN Y & WUNDERINK RG. 2018. MERS, SARS and Other Coronaviruses as Causes of Pneumonia. Respirol 23(2): 130-137. https://doi.org/10.1111/resp.13196.
    » https://doi.org/10.1111/resp.13196
  • ZHOU F ET AL. 2020. Clinical Course and Risk Factors for Mortality of Adult Inpatients with COVID-19 in Wuhan, China: A Retrospective Cohort Study. Lancet 395(10229): 1054-1062. https://doi.org/10.1016/s0140-6736(20)30566-3.
    » https://doi.org/10.1016/s0140-6736(20)30566-3
  • ZHOU Y ET AL. 2015 Protease Inhibitors Targeting Coronavirus and Filovirus Entry. Antiviral Res 116(April): 76-84.

Publication Dates

  • Publication in this collection
    05 Dec 2022
  • Date of issue
    2022

History

  • Received
    29 Nov 2021
  • Accepted
    08 Mar 2022
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br