Your new experience awaits. Try the new design now and help us make it even better

SYSTEMATIC REVIEW article

Front. Med., 09 June 2025

Sec. Nephrology

Volume 12 - 2025 | https://doi.org/10.3389/fmed.2025.1568943

This article is part of the Research TopicChallenge to the Future: Advances in post-COVID-19 intensive care NephrologyView all 4 articles

Ascertaining the mechanistic etiology of COVID-associated glomerulonephritis: a systematic review


Brendan M. CoyneBrendan M. Coyne1Danielle ItoDanielle Ito2Anam TariqAnam Tariq1Susie Q. LewSusie Q. Lew1Jeffrey KoppJeffrey Kopp3Patricia Centron VinalesPatricia Centron Vinales1Fahim MalikFahim Malik4Patrick E. Gipson*Patrick E. Gipson1*Ehsan NobakhtEhsan Nobakht1
  • 1George Washington School of Medicine and Health Sciences, Washington, DC, United States
  • 2Department of Biological Sciences, University of California, Irvine, Irvine, CA, United States
  • 3National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, United States
  • 4Washington Nephrology Associates, Rockville, MD, United States

Background: Since its first reported case in December 2019, COVID-19 disease, caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), evolved into a major pandemic throughout the world. Although COVID-19 is most often characterized as a respiratory pathology, there are also extensive reports of renal complications, such as glomerulonephritis (GN). However, the precise nature of COVID-associated glomerulonephritis (COVID-GN) has yet to be fully understood. This review seeks to elucidate COVID-GN pathophysiology by conducting an exhaustive systematic review.

Methods: Herein, we compare the different GN subtypes associated with COVID-19 in the literature. We also review the cytokines, antibodies, and genes most implicated in COVID-GN.

Results: The GN subtype with the highest number of cases associated with COVID-19 infection was focal segmental glomerulosclerosis, specifically the collapsing morphology. Meanwhile, the highest number of cases associated with COVID-19 vaccination was IgA nephropathy. The most prevalent mechanism in the literature for COVID-GN involves a cytokine storm, which may be accompanied by immune complex deposition.

Discussion: Both infection and vaccination from SARS-CoV-2 can induce robust CD4+ T cell responses promoted by an IL-6 amplifier loop of inflammation. This immune response is likely further enhanced by interactions with complement systems and the renin-angiotensin-aldosterone system (RAAS). SARS-CoV-2-mediated pathways of both direct cytotoxicity and stimulation of polyclonal immunoglobulin may converge to cause glomerular inflammation and injury. Further investigation of these inflammatory pathways may provide insight into COVID-19 pathophysiology, treatment, and long-term outcomes.

1 Introduction

Since its first reported cases of human infection in December 2019, the severe acute respiratory coronavirus 2 (SARS-CoV-2) pathogen has spread throughout every major country in the world (1). This caused a global health crisis and pandemic, with at least 81 million reported cases and over 1,777,000 deaths within the first year alone (2).

Severe acute respiratory coronavirus 2, similar to other viruses of the Coronaviridae family, enters host cells via binding to angiotensin-converting enzyme (ACE)-2 receptors and subsequent endocytosis and/or membrane fusion of the receptor-virus complex (3, 4). Binding to ACE-2 is dependent on SARS-CoV-2’s surface spike protein (S), which is proteolytically activated by host transmembrane protease serine 2 (TMPRSS2) (5). SARS-CoV-2’s ribonucleic acid (RNA) genome is replicated and translated within the cytoplasm forming new SARS-COV-2 virions (6). Furin-mediated processing of S on the virion’s surface is followed by release of the newly-formed viruses into the extracellular environment (6). This viral life cycle perpetuates throughout the body, targeting vital organs (7).

Severe acute respiratory coronavirus 2 infection can cause COVID-19, which is most often characterized as a respiratory disease (8). However, renal involvement and substantial acute kidney injury (AKI) may also occur (9). A high proportion of hospitalized COVID-19 patients present with AKI, proteinuria, and/or hematuria, suggesting glomerular or tubulointerstitial involvement (10).

Coronavirus disease 2019’s connection to renal tubular damage is well-established and comprises the majority of COVID-related AKI (9). However, glomerulonephritis (GN) is also an important complication, and its association with SARS-CoV-2 remains to be fully understood. GN appears to be one of the most destructive renal pathologies in COVID-19; COVID-19 vaccine-associated GN had higher fatality than either vaccine-associated AKI or tubulointerstitial nephritis (TIN) (11). However, there remains much uncertainty regarding the precise nature of this relationship. There is some doubt as to whether the perceived association between SARS-CoV-2 and GN is causal versus coincidental. Thus, ongoing surveillance of COVID-associated GN (COVID-GN) would be prudent as SARS-CoV-2 and its variants continue to circulate throughout the world.

2 Methods

A comprehensive review with a timeline from 1 January 2020 to 31 December 2023 was conducted in accordance with Preferred Reported Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines (12).

2.1 Study selection

A digital research database search was performed with the following key terms: COVID-19, SARS-CoV-2, glomerulonephritis, nephropathy, and nephritis. Literature was searched within four electronic databases (PubMed, MEDLINE, EBSCO, Scopus, and Google Scholar) during the time frame of 1 January 2020 through 31 December 2023.

Authors BC and DI independently screened studies by title, abstract, and full text. Relevant literature included case reports and series, observational/cohort studies, and meta-analyses. Systematic reviews, editorials, conference abstracts, experimental studies on animal models, and articles without full-text or original data available were excluded. Additional exclusion criteria included: lack of evidence of glomerulonephritis on labs or biopsy, studies describing an unspecified glomerulonephritis, and non-English articles without official translations.

2.2 Data extraction

Investigators BC and DI independently compiled key data from the included articles in a standardized Excel spreadsheet. Extracted data was then validated by all authors. Information was collected regarding author lists, publication year, country or region where the study was conducted, disease course, clinical features (e.g., serology, kidney pathology), and outcomes (i.e., morbidity, mortality, complications).

The number was tallied for cases of IgA nephropathy, anti-glomerular basement membrane disease, membranous nephropathy, C3 glomerulonephritis, ANCA-associated vasculitis and glomerulonephritis, lupus nephritis, focal segmental glomerulosclerosis, and minimal change diseases that had a temporal association with SARS-CoV-2 infection or immunization (Figure 1). Each case was further categorized by the presentation, as either acute, de novo GN versus a flared relapse of underlying GN.

FIGURE 1
www.frontiersin.org

Figure 1. (A) Tallied cases of glomerulonephritis (GNs) associated with coronavirus disease 2019 (COVID-19) infection. The highest number of reports was de novo FSGS with 196 cases. The lowest frequency was relapsed MCD with 0 reported cases. (B) Tallied cases of GNs associated with COVID-19 vaccination. The highest number of reports was relapsed IgAN with 141 cases. The lowest frequency was acute and relapsed C3GN and relapsed anti- GBM with 0 reported cases. FSGS, focal segmental glomerulosclerosis; MCD, minimal change disease; IgAN, IgA nephropathy; MN, membranous nephropathy; AAV-GN, ANCA, associated vasculitis and glomerulonephritis; LN, lupus nephritis; anti-GBM, anti-glomerular basement membrane nephritis; C3GN, C3 glomerulonephritis.

3 Results

3.1 Antibody and immune complex formation

It is well-established that SARS-CoV-2 and its vaccines can elicit robust cell-mediated and antibody-mediated immune responses (13). This can induce immune dysregulation, manifesting as non-specific immune activation and/or incitation or exacerbation of autoimmune state (1417). This immune dysregulation appears to be a core mechanism in COVID-GN (Figure 2). The precise means by which this develops remains unclear, although some potential avenues have been explored, such as SARS-CoV-2-mediated cross-reactivity in antibodies and T cells (1821).

FIGURE 2
www.frontiersin.org

Figure 2. Pathophysiologic summary of coronavirus disease (COVID)-associated glomerulonephritis (GNs). Green boxes represent primarily adaptive immune processes, yellow boxes represent primarily innate immune responses, blue boxes are neutral, red boxes indicate GN subtypes. Mechanisms that are more well-established in the literature are represented by solid arrows, whereas mechanisms that are less well-understood are represented by dashed arrows. IFN-y, interferon gamma; NF-kB, nuclear factor kappa B; TNF-a, tumor necrosis factor a; EGFR, epidermal-like growth factor receptor; STAT3, signal transducer and activator of transcription 3; ORF7a, open reading frame 7a; IL-6, interleukin 6; IL-6Ra, interleukin 6 receptor alpha; TLR, toll-like receptor; ACE-2, angiotensin-converting enzyme 2; ADAM 17, disintegrin and metalloproteinase 17; MHC, major histocompatibility complex; HLA-DR, human leukocyte antigen-DR; THSD7A, thrombospondin type-1 domain- containing 7A; NELL1, nerve epidermal growth factor- like antigen 1; Gd-lgA1, galactose-deficient lgA1; MPO, myeloperoxidase; PR3, proteinase 3.

3.1.1 IgA nephropathy

One of the common types of GN investigated is IgA nephropathy (IgAN). COVID-19 disease has shown to potentially evoke and/or exacerbate IgA vasculitis and IgAN (2244). COVID-19 vaccines have also been associated with both acute IgAN (16, 36, 4568) and flares of pre-existing IgAN (53, 57, 6989).

IgA nephropathy represents an autoimmune state that classically follows mucosal infections, in which IgA is upregulated by the immune system and can subsequently deposit into tissues such as the glomerular mesangium (90). Given that SARS-CoV-2 antigens act on receptors widely expressed throughout respiratory and GI mucosa (3), it is quite conceivable how IgAN may follow COVID-19 infection or immunization.

It is possible that COVID-19 exposure activates B cells to produce especially high levels of IgA, which form acute immune complexes that deposit in glomeruli (22, 91). Indeed, the first immunoglobulin detected in COVID-19, anti-SARS-CoV-2 IgA, precedes both IgM and IgG serology (92). This may implicate IgA as one of the predominant agents mediating COVID-19 glomerular damage. However, it is debated as to whether this is attributed to acute IgA production or pre-existing IgA deposits in the glomerular mesangium that are simply “unmasked” by SARS-CoV-2 (70, 74).

Of note, some COVID-associated IgAN cases present with elevated galactose-deficient-IgA1 (Gd-IgA1) in the serum or Gd-IgA1 mesangial deposits (65, 93). Mesangial deposition of Gd-IgA1, an IgA variant caused by aberrant glycosylation, is believed to play a role in IgAN (94). Recognition of SARS-CoV-2 antigens by Gd-IgA1 may result in immune complex and complement deposition in the glomerular mesangium (93). Future research on the induction of Gd-IgA1 and anti-glycan immune complexes may provide insight into these processes.

3.1.2 Anti-glomerular basement membrane disease

Another autoimmune nephropathy associated with COVID-19 is anti-GBM disease (also known as Goodpasture syndrome). Certain cases of COVID-19 disease or COVID-19 vaccines may be associated with anti-GBM nephritis, usually presenting with crescentic, rapidly progressive glomerulonephritis (16, 41, 44, 52, 55, 74, 95105). This is correlated by a significantly increased incidence of anti-GBM disease since the start of the COVID-19 pandemic (106).

Goodpasture syndrome is classically described as an autoimmune disorder driven by molecular mimicry and autoantibodies (107). Similarly to Streptococcus- and influenza-induced anti-GBM nephritis, the severe endothelial damage from SARS-CoV-2 could conceivably expose basement membrane antigens (e.g., Goodpasture antigen) in the respiratory tract and renal tissue; epitope similarity would then cause autoantibodies to attack glomeruli (96, 108). However, given that the literature remains sparse, the causal relationship between COVID-19 and anti-GBM disease remains speculative.

3.1.3 Membranous Nephropathy

Severe acute respiratory coronavirus 2 infection has been implicated in potentially inducing or exacerbating primary and secondary membranous nephropathy (MN) (35, 44, 96, 109111). Additionally, immunization with COVID-19 vaccines was associated with both acute (36, 47, 48, 52, 53, 55, 112117) and relapsed MN (16, 78, 82, 83, 118, 119).

Primary MN often results from autoimmune activity, usually caused by immune complex deposition and presenting with autoantibodies against phospholipase A2 receptor (PLA2R), although autoantibodies against thrombospondin type-1 domain-containing 7A (THSD7A) and nerve epidermal growth factor-like antigen 1 (NELL1) have been implicated as well (120, 121). These antibodies form immune complexes which deposit in sub-epithelial tissue of glomeruli, causing basement membrane thickening (122). Although classically associated with primary MN, PLA2R serologies have also been detected in secondary MN with viral etiology (e.g., hepatitis B, hepatitis C) (120).

It is possible that both primary and secondary MN associated with SARS-CoV-2 involve virus-evoked anti-PLA2R complexes (123). Although some cases test positive for THSD7A or NELL-1 (16, 113), the majority are PLA2R+ (16, 109, 118). Seeing as how PLA2R is expressed in the respiratory tract, SARS-CoV-2-mediated damage may release PLA2R from respiratory epithelial cells, triggering the development of autoantibodies that attack glomerular tissue (123).

However, COVID-19 vaccine-associated MN cases have also tested positive for anti-PLA2R in the serum and/or renal tissue (16, 36, 47, 48, 52, 78, 112, 114, 118). This suggests that direct respiratory damage from SARS-CoV-2 virus is not necessarily required to induce PLA2R+ MN. Rather, messenger RNA (mRNA) or surface antigen may somehow diminish immune tolerance for PLA2R antigen (118), but this hypothesis requires further evidence.

3.1.4 C3 Glomerulonephritis

There are some cases reporting a potential association between SARS-CoV-2 infection and C3 glomerulonephritis (C3GN) (124126). C3GN is a pathology formerly categorized as a subtype of membranoproliferative glomerulonephritis (MPGN), which is now used primarily to describe glomerular injury patterns as opposed to a specific disease diagnosis (127, 128). A process closely related to C3GN, immune complex MPGN (IC-MPGN) has also been reported to follow SARS-CoV-2 infection or vaccination (35, 47, 78, 124, 125, 129, 130). One case exhibited lupus-like features with a “full house” pattern on immunofluorescence (129). Such findings are comparable to the lupus-like biopsies in human immunodeficiency virus (HIV)-associated immune complex kidney disease (HIVICK), which is similarly common among White patients (131).

In HIVICK, it is proposed that systemic and intrarenal immune activity triggers polyclonal B cells and hypergammaglobulinemia with resultant glomerular hyperplasia (132, 133). This mechanism may similarly underlie the strong IgG and IgM staining that was observed in a patient with COVID-19 and C3GN (126). This was correlated with the eventual development of tubuloreticular inclusions (“interferon footprints”), possibly indicating a strong contributory role of interferon (IFN) cytokines (126).

However, research in this area is limited, and there does not seem to be substantial evidence that COVID-19 can trigger C3GN. It is important to recognize that neither C3GN nor IC-MPGN demonstrate consistent, strong associations with COVID-19. However, identification of their pathophysiology may guide researchers toward which mechanisms are more likely or less likely underpinning COVID-GN.

3.1.5 Vasculitis-associated glomerulonephritis

Coronavirus disease 2019 has been previously associated with autoimmune destruction of glomerular blood vessels, which can cause crescentic, necrotizing GN with fibrinoid necrosis (134, 135). Biopsies typically stain negative or very low for immune complexes; hence the disease is often referred to as Pauci-immune GN (136).

Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis glomerulonephritis (AAV-GN) is a group of disorders sharing similar features on biopsy (e.g., often crescentic, Pauci-immune) and classically driven by autoantibodies against neutrophil proteins. Variations of AAV-GN include granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (formerly Churg-Strauss syndrome).

Coronavirus disease 2019 disease has been associated with new-onset GPA (137139), relapsed GPA (140), and new-onset MPA (141). COVID-19 vaccination has also been temporally linked to new-onset GPA (142, 143), and both new-onset and relapsed MPA (82, 144, 145). Other variations of AAV-GN have also developed following SARS-CoV-2 infection and immunization (16, 17, 36, 38, 44, 47, 48, 55, 78, 99, 100, 146180).

The hypothesized mechanism for COVID-associated AAV-GN involves COVID-triggered immune dysregulation; specifically, it is possible that SARS-CoV-2 proteins promote the development of autoantibodies against neutrophil proteins such as myeloperoxidase (MPO) and proteinase 3 (PR3), which is characteristic of classic AAV-GN (178, 180182). The pathophysiology of AAV strongly implicates the role of neutrophil extracellular traps (NETs) as sources of autoantigens, presenting MPO and PR3 to adaptive immune cells (183). NETs normally play a significant role in host defense, but NET dysregulation can lead to angiopathy and ANCA development (184). NETs have been implicated as a critical player in SARS-CoV-2 infection and the cytokine storm that can follow (185, 186); they are also present in kidney biopsies of some COVID-19 patients (187). Autoantigen presentation by NETs is thus a possible mechanism for COVID-associated AAV-GN (178, 188).

The reports of both c-ANCA (45, 189194) and p-ANCA (16, 52, 53, 84, 101, 176, 195208) GN following mRNA vaccination potentially suggests that SARS-CoV-2-associated NETs and AAV-GN are mediated by the virus’s S protein, or even the viral mRNA itself. Indeed, mRNA detection by dendritic cells and macrophages leads to increased activity of type 1 interferon (IFN-1) and other cytokines, which prime neutrophils to release reactive oxygen species and lytic enzymes, and facilitate the formation of NETs (Figure 3) (209).

FIGURE 3
www.frontiersin.org

Figure 3. Proposed mechanism of the IL-6 amplifier pathway in the setting of coronavirus disease 2019 (COVID-19). Ang II, angiotensin II; ACE-2, angiotensin-converting enzyme 2; ATR1, angiotensin II type 1 receptor; ADAM17, disintegrin and metalloproteinase 17; TNF-a, tumor necrosis factor alpha; EGFR, epidermal-like growth factor receptor; NF-kB, nuclear factor kappa B; STAT3, signal transducer and activator of transcription 3; IL-6, interleukin 6; IL-6Ra, interleukin 6 receptor alpha; IFN-1, type 1 interferon; IFN-y, interferon gamma; TLR, toll-like receptor; NET, neutrophil extracellular traps; MPO, myeloperoxidase; PR3, proteinase 3; Gd-IgA1, galactose-deficient IgA1.

In addition to NET dysregulation, abnormal expression and activity of major histocompatibility complex (MHC) class II molecules is also implicated in AAV-GN pathogenesis, as outlined in Figure 4 (146, 184, 210214).

FIGURE 4
www.frontiersin.org

Figure 4. Proposed mechanism of MHC-II contribution to coronavirus disease (COVID)-associated AAV-GN. MON, monocyte; Mo, macrophage; CYT, pro-inflammatory cytokines; CD4 T, CD4+ T cell; CD8 T, CD8+ T cell; MHC II, major histocompatibility complex class II; TCR, T cell receptor; AAV-GN, ANCA- associated vasculitis and glomerulonephritis; HLA-DR, human leukocyte antigen receptor; MPO, myeloperoxidase; PR3, proteinase 3.

3.1.6 Lupus nephritis

Whether through clinical diagnosis or confirmed by biopsy, there have been a number of reports correlating SARS-CoV-2 infection with new-onset lupus nephritis (LN) (44, 215222) as well as flares of chronic LN (96, 223226) with or without systemic lupus erythematosus (SLE). Additionally, adenoviral vector and mRNA vaccines against SARS-CoV-2 have been associated with de novo (47, 52, 55, 78, 227, 228) and relapsed (48, 82, 229232) LN and SLE. LN biopsies in COVID-19 are consistent with classic LN, which generally stains positive for deposits with IgG, C3, and C1q dominance (233). Such clinical and morphological findings contribute to the growing evidence of COVID-19 exacerbating underlying autoimmunity.

3.2 Cytokine-mediated glomerular damage

While an effective immune response to SARS-CoV-2 involves both B- and T cells, the rapid nature of GN onset post-COVID-19 vaccination implicates T cells as the more important mediators of glomerular damage (234). T cells respond to viral mRNA with rapid production of cytokines (e.g., IFN-gamma, tumor necrosis factor (TNF)-alpha, interleukin (IL)-6), which can both directly damage glomeruli as well as augment B cell activity and immune complex deposition (Figure 3) (234).

3.2.1 Minimal change disease

Coronavirus disease 2019 has been correlated with podocytopathies that traditionally were thought to be driven by non-immune complex deposition processes. For example, COVID-19 infection has been associated with minimal change disease (MCD), a nephrotic syndrome previously thought to be cytokine-mediated (33, 35, 36, 43, 44, 96, 125, 235237). Vaccination against the virus also may develop new-onset or relapsed MCD (16, 36, 47, 48, 52, 53, 55, 78, 82, 83, 237258). While recent emerging evidence suggests that a number of MCD cases may in fact be driven by pathologic antibodies to nephrin (259), in the majority of cases, there remains a complex picture of interactions which likely reflect multiple parts of the immune system, including cytokine activity, T cell dysregulation, and B cell activation (260).

3.2.2 Focal segmental glomerulosclerosis

Focal segmental glomerulosclerosis (FSGS) may similarly be driven in large part by cytokine damage rather than immune complex deposition. Additionally, recurrence or relapse of FSGS is thought to be potentially mediated by an unidentified circulating factor, whether it be an unidentified antibody, T cell cytokine, or non-immunogenic permeability factor, which could potentially be elicited by viruses such as SARS-CoV-2, leading to podocyte toxicity (Figure 2) (261, 262). FSGS has developed following the immune response elicited by SARS-CoV-2 infection (23, 44, 263270) and vaccination (36, 52, 55, 78, 82, 271274).

Collapsing FSGS (cFSGS), also known as collapsing glomerulopathy, is a morphological variant of FSGS. A plethora of cases have identified a temporal link between COVID-19 and cFSGS, particularly in the African-American population, with a higher incidence and poorer prognosis among high-risk apolipoprotein L1 (APOL1) variants (36, 37, 55, 125, 222, 235, 237, 271, 275303).

Coronavirus disease-associated cFSGS has incited many comparisons to HIV-associated nephropathy (HIVAN). HIVAN appears to have a similar genetic predisposition, with higher incidence among African-Americans carrying APOL1 variants (304). More recently, non-HIV viruses, including hepatitis C virus (HCV) (305) and SARS-CoV-2 (304), have been implicated in its pathogenesis. Given their notable similarities, it may be fruitful to apply clinical knowledge of HIVAN pathogenesis and management toward COVID-associated cFSGS. Based on previous glucocorticoid use in HIVAN, one group of authors included prednisone in their treatment regimen for COVID-associated cFSGS, yielding successful outcomes (306). Such approaches may be useful moving forward, given that treatment for COVID-associated cFSGS is generally challenging with a poor prognosis (302).

It is important to note that whereas HIVAN is primarily driven by direct viral invasion and cytopathy of renal cells, this does not seem to be the case for COVID-associated FSGS (307). Many investigators propose that both collapsing and non-collapsing glomerulopathy in the setting of COVID-19 result from cytokine-mediated activation of innate and adaptive immune cells (263, 275, 282, 308). IFN pathways may be particularly important in COVID-associated cFSGS, as demonstrated by the presence of “IFN footprints” in some patients (96, 308). Previous research shows that IFN therapy may even induce cFSGS and other nephropathies (309, 310).

It is possible that there is a convergence in cytokine pathways between COVID-19 and other inflammatory states, such as in transplant immunity. Acute T cell-mediated allograft rejection has developed subsequent to SARS-CoV-2 infection and immunization, correlated with a CD3 + T cell predominance (96, 226, 237, 263). Furthermore, both IFN and granulocyte colony-stimulating factor–which are elevated in COVID-19 (311313)–are important cytokines for T cell-mediated exacerbation of autoimmune GN (314) and acute transplant rejection (315).

Multiple allograft recipients have developed collapsing glomerulopathy in the setting of COVID-19 infection and immunization, with two patients expressing low-risk APOL1 alleles (48, 226, 263, 264, 279, 295, 316321). Similarly, non-transplant patients have also presented with COVID-associated cFSGS without high-risk APOL1 polymorphisms; these patients had significant pre-existing conditions, including SLE and HIV (222, 291). It is possible that a pre-existing state of immune dysregulation (e.g., SLE, HIV, transplant immunity) may predispose or “prime” cytokine pathways to be triggered by COVID-19, resulting in cFSGS even in the absence of high-risk APOL1 mutations. Still, the presence of a high-risk APOL1 variant likely confers a worse prognosis, as exemplified by a high-risk APOL1 patient developing severe COVID-19 infection and cFSGS despite having prior immunity to SARS-CoV-2 (316).

Although the nephrotoxic effect of high-risk APOL1 variants is not fully understood, some proposed mechanisms include impaired endolysosomal trafficking, inflammasome activation, and APOL3 control of actomyosin in podocytes (322). The inflammatory response to COVID-19 may act as a “second hit” for APOL1 variants that at baseline are at high risk for developing glomerulopathy (275). Previously established second-hit triggers of glomerulopathies in high-risk APOL1 variants include HIV, SLE, and IFN therapy (323). SARS-CoV-2 is being considered as a second-hit trigger for cFSGS in a similar manner.

Innate immune pathways (e.g., IFN-1) can enhance APOL1 expression, and thus may facilitate COVID-associated cFSGS via upregulation of high-risk APOL1 alleles (Figure 2) (324326). However, some cases of COVID-associated cFSGS in high-risk APOL1 variants did not exhibit IFN footprints (282, 285) or elevated IFN-1 expression (275). Additionally, Meliambro et al. did not find significant differences in APOL1 expression between controls and a high-risk APOL1 COVID-19 patient with c-FSGS (327). However, this may have been confounded by the anti-inflammatory action of hydroxychloroquine or the delayed kidney biopsy (327).

Overall, the literature demonstrates that COVID-associated cFSGS can develop in low, intermediate, or high-risk APOL1 individuals, with greater frequency and severity in the latter group and especially affecting those of African ancestry. Although IFN pathways are potentially implicated, knowledge gaps remain regarding the specific cytokines most strongly contributory to the glomerular injury of COVID-associated FSGS.

4 Hypothesized pathophysiology of COVID-associated glomerulonephritis

4.1 Controversies surrounding direct viral damage versus immune-mediated damage

Coronavirus disease-GN may arise via a variety of potential mechanisms. These include, but are not limited to: direct viral cytopathy, immune hyperactivation, hemodynamic instability, and metabolic imbalances (9). Researchers have attempted to tease out which underlying mechanisms contribute the most.

The first reported renal biopsy of a live COVID-19 patient showed collapsing glomerulopathy without signs of viral presence within renal cells (276). Many subsequent COVID-GN reports since then have similarly demonstrated a low likelihood that direct viral cytotoxicity is responsible for COVID-GN (27, 96, 275, 277, 278, 280, 328).

This may be the result of renal ACE-2 and TMPRSS2 expression. ACE-2 seems to be predominant in proximal tubules, whereas TMPRSS2 appears to be predominant in the distal tubules (329, 330). Given that ACE-2 and TMPRSS2 work in tandem with one another, their differential expression in separate regions of the nephron may limit SARS-CoV-2’s ability to enter.

Still, some authors maintain that SARS-CoV-2 infiltration into renal cells may cause COVID-GN via direct cytotoxicity and vasculitis (331, 332). Several cases observed particles within tubular epithelium and podocytes possibly suggestive of SARS-CoV-2; however, these particles may instead be clathrin-coated vesicles that are normally found within cells (22, 23, 235, 281, 318, 319, 333).

Some articles have also reported SARS-CoV-2 mRNA within kidney tissue (20, 96, 334). However, other biopsies have failed to demonstrate this (27, 110, 129, 277). Additionally, a number of COVID-GN cases occur post-infection, after viral clearance and negative PCR testing (17, 129, 138). COVID-GN has also developed following mRNA, adenoviral-vector, and inactivated vaccines, further demonstrating that direct viral infection is not required for nephropathy (47).

A final piece of evidence dissuading against direct virus-mediated COVID-GN is the presentation pattern. COVID-19 is associated with a wide spectrum of glomerular and tubular disease states, a pattern more consistent with systemic immune hyperactivation as opposed to direct viral cytotoxicity (96, 187). Overall, it appears unlikely that direct virus-mediated damage is predominant in COVID-GN.

4.2 Cytokine storm and IL-6 amplifier

Severe acute respiratory coronavirus infection is strongly associated with upregulated activity of CD4+ and CD8+ T cells, despite a decrease in the absolute number of CD8+ T cells (335, 336). COVID-19 vaccines can also stimulate robust CD4+/CD8+ responses with enhanced cytokine production (19, 337, 338). Furthermore, it has been established that such T cell dysregulation and hyperstimulation is linked to both immune complex-driven GN and cytokine-driven GN, including IgAN, LN, MCD, and FSGS (339341).

We stress that a major pillar of COVID-GN is driven by cytokines, specifically a T cell-mediated cytokine storm. As summarized in Figure 3, current research implicates the IL-6 amplifier system in propagating the cytokine storm of COVID-19 (13, 337, 342346). The age-dependent strength of the IL-6 amplifier’s feedback loop may correspond with the age-dependent increase in COVID-19 morbidity and mortality (347).

Interleukin-6-mediated lymphocyte recruitment may play a role in the humoral and cell-mediated processes underlying COVID-GN. Studies have found elevated IL-6 levels in the serum of patients with COVID-related IgAN and vasculitis (25, 27). Furthermore, IL-6 may strongly alter IgA glycosylation machinery and significantly contribute to glomerular IgA deposition (348350). It is possible that the IL-6 amplifier provoked by SARS-CoV-2 promotes the production of Gd-IgA1, resulting in complex deposition within the glomeruli. IFN-1 cytokines, specifically IFNa, may further contribute to COVID-associated flares of chronic IgAN (351, 352). Increased phosphorylation and activation of STAT3 has also been observed in COVID-GN (327). Targeting the expression or activation of factors like STAT3, IFN-1, and IL-6 may hold therapeutic potential for COVID-GN.

It is likely that the IL-6 feedback loop and cytokine storm in COVID-19 are enhanced by complement pathways. Complement activation has shown to contribute to SARS-CoV-2 pathogenesis and severity (353). COVID-19 patients have elevated serum levels of complement proteins like C5a, which can boost IL-6 and TNFa expression (353355). Similarly, COVID-GN can also present with elevated C5a levels, increased C5aR activity, and decreased C3/C4 levels (356). It is thus possible that complement pathways propagate COVID-GN via potentiation of cytokine activity.

Overall, although the exact relationship remains unclear, there is mounting evidence that COVID-GN involves an IL-6 amplifier-driven hyperinflammatory storm.

4.3 SARS-CoV-2 superantigen

Cheng et al. (357) found that SARS-CoV-2’s S protein has superantigen characteristics, exhibiting high affinity for complementarity determining regions of both α- and β-chain variable domains of T cell receptors (TCRs), and that this interaction is further strengthened by certain mutations in the virus’s genome.

Researchers have also found that there is a skew in expression of TCR repertoire among COVID-19 patients with hyperinflammation (358). These effects seem to be due to a sequence motif within S’s binding epitope that is unique to SARS-CoV-2; this motif is absent in other coronaviruses and instead resembles bacterial superantigens in both sequence and structure (358). Enterotoxin B and C of staphylococci can act as superantigens, inducing GN via mass stimulation of T cell cytokines and polyclonal immunoglobulin (359, 360). SARS-CoV-2’S protein may act as a superantigen in a similar manner, driving the development of GN via a cytokine storm.

4.3 Interactions with the renin-angiotensin-aldosterone system

Interactions with RAAS may contribute to the cytokine storm in COVID-19. Binding of SARS-CoV-2 to ACE-2 results in increased angiotensin II (Ang II) binding to angiotensin II type 1 receptor (ATR1), resulting in the induction of disintegrin and metalloproteinase 17 (ADAM17), which not only generates the mature forms of epidermal growth factor receptor (EGFR) and TNF-a, but also processes the membrane form of IL-6 receptor alpha (IL-6Ra) into its soluble form (3, 361363). The net downstream effect is IL-6 amplifier activation (Figure 3). COVID-GN thus may involve dysregulation of the ACE-2 and Ang II axis.

However, conflicting findings have been reported, summarized in Table 1 (346, 364375). Interestingly, in one COVID-19 patient with cFSGS, there was no significant difference in ACE-2 transcript expression compared to normal kidney samples (327). However, translational and post-translational modifications of ACE-2 can cause discrepancies between transcript and protein expression (371, 376).

TABLE 1
www.frontiersin.org

Table 1. Opposing views with seemingly conflicting evidence in the literature regarding the role of angiotensin-converting enzyme-2 (ACE-2) expression and renin-angiotensin-aldosterone system (RAAS) in the development and severity of coronavirus disease-associated glomerulonephritis (COVID-GN).

There is a shortage of conclusive literature, but the current evidence seems to suggest that decreased ACE-2 expression predisposes patients to severe COVID-19 and COVID-GN. Regardless, ACE-2 receptors should be further investigated as a potentially crucial component of COVID-GN pathogenesis.

5 Discussion and conclusion

The overall literature seems to suggest that there may be an association between COVID-19 and both de novo and relapsed GN. The most common overall type of COVID-GN was FSGS, particularly cFSGS. In infection, the most common cases were de novo FSGS followed by de novo AAV-GN. In vaccination, the most common cases were relapsed IgAN followed by de novo AAV-GN. IgAN had the most relapse cases in both infection and vaccination, potentially suggesting a greater prominence of “unmasking” mechanisms for COVID-associated IgAN.

Prognosis seems to differ between different GN pathologies–patients who developed COVID-associated IgAN or MCD were generally more likely to recover kidney function with appropriate management (55).

The numerous instances of mixed glomerular histologies (e.g., MN and cFSGS, IgAN, and AAV-GN) potentially suggests a multifactorial pathophysiology for COVID-GN (37, 49, 55, 60). Cytokine storms, which may or may not be accompanied by immune complexes, is the most consistently prevalent mechanism described within the literature. SARS-CoV-2 infection and immunization likely induce robust CD4+ upregulation, ensuing the IL-6 amplifier, which is enhanced by RAAS and complement pathways. Cytokines of interest include IL-6, IFN-1, and TNFa, which may mediate polyclonal immunoglobulin stimulation and/or direct cytotoxicity of glomeruli. Targeting these pathways may prove fruitful in our understanding of COVID-19 disease and treatment.

Genetic interactions with SARS-CoV-2’s inflammatory cascade can predispose certain individuals to developing severe COVID-19 and GN. Mutations and polymorphisms in ATR1, ACE-2, and APOL1 should be further investigated as potential predisposing factors for COVID patients.

There was a higher number of GN cases associated with COVID-19 vaccines than infections in the literature (Figure 1). However, it is unlikely that vaccination confers a greater risk of GN. Vaccine cases are generally more controlled environments than active COVID-19, making them easier to study and report. Furthermore, multiple large population-wide studies across different countries have found that the COVID-19 vaccine rollout did not significantly increase overall incidence of glomerular disease (377, 378). Among the different types of COVID-19 vaccines, mRNA vaccines were the most commonly reported in association to GN. This does not necessarily indicate that mRNA vaccines inherently carry a higher propensity for GN, but rather, likely reflects the widespread availability of mRNA vaccines for COVID-19 across the global population.

Although this review has aimed to be fully comprehensive, there are limitations. Due to the retrospective nature intrinsic to systematic reviews, we are unable to definitively claim a causative effect between COVID-19 and GN. There is a possibility that some COVID-GN cases were due to concurrent disease or incidental timing. For example, one case of AAV, not included in this review, was initially believed to be secondary to COVID-19 vaccination, until underlying Dengue infection was identified (379, 380). This review also includes both case reports and national or international registries, resulting in a slight theoretical possibility of “double-counted” cases.

Given the novel nature and ever-developing literature surrounding SARS-CoV-2 pathophysiology, it is not surprising that COVID-GN presents with unsettled results. For example, precise etiology is difficult to ascertain in dual diagnosis cases (e.g., AAV-GN and anti-GBM) (99). It can also be difficult to distinguish new-onset AAV in COVID-19 patients, due to pulmonary and vasculitis-like symptomatology sometimes resembling one another (381384).

It was unclear if certain COVID-GN cases were acute or chronic processes. Some patients had a prior history of microscopic hematuria, yet due to their subclinical nature, they were never biopsied to confirm a GN diagnosis (60, 80). Upon exposure to SARS-CoV-2 infection or immunization, they developed new-onset gross hematuria and/or AKI, prompting a renal biopsy, leading to a GN diagnosis (60). This review tallied such cases as relapses–although the GN diagnosis was new, the patient history suggested an “unmasking” of underlying pathology.

This review specifies particular T cell types, antibodies, cytokines, and genes as requiring further investigation. However, other avenues (e.g., regulatory T cell inhibition) are also underexplored mechanisms for COVID-GN. The pathogenesis of COVID-GN is most likely multifactorial, involving multiple pathways of the immune system, possibly with secondary systemic effects (e.g., hypoxia).

This review is restricted by the retrospective evaluation of the limited number of cases published between 2020 and 2023. Continued attention toward COVID-GN may elucidate greater facets of its pathophysiology and treatments.

Data availability statement

The original contributions presented in this study are included in this article/supplementary material, further inquiries can be directed to the corresponding author.

Author contributions

BC: Conceptualization, Methodology, Data curation, Investigation, Writing – review and editing, Writing – original draft, Visualization. DI: Data curation, Investigation, Methodology, Writing – original draft. AT: Supervision, Validation, Visualization, Writing – review and editing. SL: Supervision, Validation, Writing – review and editing. JK: Writing – review and editing, Validation. PV: Validation, Writing – review and editing. FM: Conceptualization, Writing – review and editing. PG: Writing – review and editing, Supervision, Validation. EN: Supervision, Writing – review and editing, Validation.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Salahshoori I, Mobaraki-Asl N, Seyfaee A, Nasirabad N, Dehghan Z, Faraji M, et al. Overview of COVID-19 disease: Virology, epidemiology, prevention diagnosis, treatment, and vaccines. Biologics. (2021) 1:2–40. doi: 10.3390/biologics1010002

Crossref Full Text | Google Scholar

2. Covid-19 deaths. Datadot. (2024). Available online at: https://data.who.int/dashboards/covid19/deaths (accessed July 20, 2024).

Google Scholar

3. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. (2020) 181:271–280.e8. doi: 10.1016/j.cell.2020.02.052

PubMed Abstract | Crossref Full Text | Google Scholar

4. Zhou P, Yang X, Wang X, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. (2020) 579:270–3. doi: 10.1038/s41586-020-2012-7

PubMed Abstract | Crossref Full Text | Google Scholar

5. Wu A, Peng Y, Huang B, Ding X, Wang X, Niu P, et al. Genome composition and divergence of the novel coronavirus (2019-nCoV) originating in China. Cell Host Microbe. (2020) 27:325–8. doi: 10.1016/j.chom.2020.02.001

PubMed Abstract | Crossref Full Text | Google Scholar

6. Murgolo N, Therien A, Howell B, Klein D, Koeplinger K, Lieberman L, et al. SARS-CoV-2 tropism, entry, replication, and propagation: Considerations for drug discovery and development. PLoS Pathog. (2021) 17:e1009225. doi: 10.1371/journal.ppat.1009225

PubMed Abstract | Crossref Full Text | Google Scholar

7. Liu J, Li Y, Liu Q, Yao Q, Wang X, Zhang H, et al. SARS-CoV-2 cell tropism and multiorgan infection. Cell Discov. (2021) 7:17. doi: 10.1038/s41421-021-00249-2

PubMed Abstract | Crossref Full Text | Google Scholar

8. Sah R, Rodriguez-Morales A, Jha R, Chu D, Gu H, Peiris M, et al. Complete genome sequence of a 2019 Novel Coronavirus (SARS-CoV-2) strain isolated in Nepal. Microbiol Resour Announc. (2020) 9:e00169-20. doi: 10.1128/MRA.00169-20

PubMed Abstract | Crossref Full Text | Google Scholar

9. Legrand M, Bell S, Forni L, Joannidis M, Koyner J, Liu K, et al. Pathophysiology of COVID-19-associated acute kidney injury. Nat Rev Nephrol. (2021) 17:751–64. doi: 10.1038/s41581-021-00452-0

PubMed Abstract | Crossref Full Text | Google Scholar

10. Cheng Y, Luo R, Wang K, Zhang M, Wang Z, Dong L, et al. Kidney disease is associated with in-hospital death of patients with COVID-19. Kidney Int. (2020) 97:829–38. doi: 10.1016/j.kint.2020.03.005

PubMed Abstract | Crossref Full Text | Google Scholar

11. Yoon S, Lee H, Kim D, Kim J, Kim Y, Moon J, et al. AKI, glomerulonephritis, and tubulointerstitial nephritis following vaccination: VigiBase analysis: Fr-or02. J Am Soc Nephrol (2023) 34:29–29. doi: 10.1681/ASN.20233411S129c

PubMed Abstract | Crossref Full Text | Google Scholar

12. Hutton B, Salanti G, Caldwell D, Chaimani A, Schmid C, Cameron C, et al. The PRISMA extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: Checklist and explanations. Ann Intern Med. (2015) 162:777–84. doi: 10.7326/M14-2385

PubMed Abstract | Crossref Full Text | Google Scholar

13. Sahin U, Muik A, Vogler I, Derhovanessian E, Kranz L, Vormehr M, et al. BNT162b2 vaccine induces neutralizing antibodies and poly-specific T cells in humans. Nature. (2021) 595:572–7. doi: 10.1038/s41586-021-03653-6

PubMed Abstract | Crossref Full Text | Google Scholar

14. Tahaghoghi-Hajghorbani S, Zafari P, Masoumi E, Rajabinejad M, Jafari-Shakib R, Hasani B, et al. The role of dysregulated immune responses in COVID-19 pathogenesis. Virus Res. (2020) 290:198197. doi: 10.1016/j.virusres.2020.198197

PubMed Abstract | Crossref Full Text | Google Scholar

15. Vlachoyiannopoulos P, Magira E, Alexopoulos H, Jahaj E, Theophilopoulou K, Kotanidou A, et al. Autoantibodies related to systemic autoimmune rheumatic diseases in severely ill patients with COVID-19. Ann Rheum Dis. (2020) 79:1661–3. doi: 10.1136/annrheumdis-2020-218009

PubMed Abstract | Crossref Full Text | Google Scholar

16. Klomjit N, Alexander M, Fervenza F, Zoghby Z, Garg A, Hogan M, et al. COVID-19 Vaccination and Glomerulonephritis. Kidney Int Rep. (2021) 6:2969–78. doi: 10.1016/j.ekir.2021.09.008

PubMed Abstract | Crossref Full Text | Google Scholar

17. Jalalzadeh M, Valencia-Manrique J, Boma N, Chaudhari A, Chaudhari S. Antineutrophil cytoplasmic antibody-associated glomerulonephritis in a case of scleroderma after recent diagnosis With COVID-19. Cureus. (2021) 13:e12485. doi: 10.7759/cureus.12485

PubMed Abstract | Crossref Full Text | Google Scholar

18. Vojdani A, Kharrazian D. Potential antigenic cross-reactivity between SARS-CoV-2 and human tissue with a possible link to an increase in autoimmune diseases. Clin Immunol. (2020) 217:108480. doi: 10.1016/j.clim.2020.108480

PubMed Abstract | Crossref Full Text | Google Scholar

19. Woldemeskel B, Garliss C, Blankson JN. SARS-CoV-2 mRNA vaccines induce broad CD4+ T cell responses that recognize SARS-CoV-2 variants and HCoV-NL63. J Clin Invest. (2021) 131:e149335. doi: 10.1172/JCI149335

PubMed Abstract | Crossref Full Text | Google Scholar

20. Puelles V, Lütgehetmann M, Lindenmeyer M, Sperhake J, Wong M, Allweiss L, et al. Multiorgan and renal tropism of SARS-CoV-2. N Engl J Med. (2020) 383:590–2. doi: 10.1056/NEJMc2011400

PubMed Abstract | Crossref Full Text | Google Scholar

21. Shah S, Danda D, Kavadichanda C, Das S, Adarsh M, Negi V. Autoimmune and rheumatic musculoskeletal diseases as a consequence of SARS-CoV-2 infection and its treatment. Rheumatol Int. (2020) 40:1539–54. doi: 10.1007/s00296-020-04639-9

PubMed Abstract | Crossref Full Text | Google Scholar

22. Pérez A, Torregrosa I, D’Marco L, Juan I, Terradez L, Solís M, et al. IgA-Dominant infection-associated glomerulonephritis following SARS-CoV-2 Infection. Viruses. (2021) 13:587. doi: 10.3390/v13040587

PubMed Abstract | Crossref Full Text | Google Scholar

23. Su H, Yang M, Wan C, Yi L, Tang F, Zhu H, et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. (2020) 98:219–27. doi: 10.1016/j.kint.2020.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

24. Hanna C, Herrera Hernandez L, Bu L, Kizilbash S, Najera L, Rheault M, et al. IgA nephropathy presenting as macroscopic hematuria in 2 pediatric patients after receiving the Pfizer COVID-19 vaccine. Kidney Int. (2021) 100:705–6. doi: 10.1016/j.kint.2021.06.032

PubMed Abstract | Crossref Full Text | Google Scholar

25. Sandhu S, Chand S, Bhatnagar A, Dabas R, Bhat S, Kumar H, et al. Possible association between IgA vasculitis and COVID-19. Dermatol Ther. (2021) 34:e14551. doi: 10.1111/dth.14551

PubMed Abstract | Crossref Full Text | Google Scholar

26. Huang Y, Li X, Li Y, Dai W, Shao T, Liu W, et al. Clinical and pathological findings of SARS-CoV-2 infection and concurrent IgA nephropathy: A case report. BMC Nephrol. (2020) 21:504. doi: 10.1186/s12882-020-02163-3

PubMed Abstract | Crossref Full Text | Google Scholar

27. Suso A, Mon C, Oñate Alonso I, Galindo Romo K, Juarez R, Ramírez C, et al. IgA vasculitis with nephritis (Henoch-Schönlein Purpura) in a COVID-19 Patient. Kidney Int Rep. (2020) 5:2074–8. doi: 10.1016/j.ekir.2020.08.016

PubMed Abstract | Crossref Full Text | Google Scholar

28. Wang B, Grand A, Schub M, Singh H, Ortiz Melo D, Howell D. Renal biopsy in systemic infections: Expect the unexpected. Ultrastruct Pathol. (2023) 47:22–9. doi: 10.1080/01913123.2022.2164099

PubMed Abstract | Crossref Full Text | Google Scholar

29. Jankowski E, Schlosser M, Wiech T, Wolf G, Busch M. SARS-CoV-2 infection: A possible trigger for the recurrence of IgA nephropathy after kidney transplantation? J Nephrol. (2023) 36:1683–7. doi: 10.1007/s40620-023-01684-y

PubMed Abstract | Crossref Full Text | Google Scholar

30. Uchida T, Sakai T, Hoshino T, Kojima A, Konno O, Yamada M, et al. Acute exacerbation of immunoglobulin A nephropathy complicated by alveolar hemorrhage after coronavirus disease 2019 vaccination: A case report. Medicine. (2023) 102:e36091. doi: 10.1097/MD.0000000000036091

PubMed Abstract | Crossref Full Text | Google Scholar

31. Elgardt I, Carmi O, Levy Y. IgA Nephropathy (Henoch-Schönlein Purpura) Associated with recent COVID-19 Infection. Isr Med Assoc J. (2022) 24:697–8.

Google Scholar

32. Oñate I, Ortiz M, Suso A, Mon C, Galindo K, Lentisco C, et al. IgA vasculitis with nephritis (Henoch-Schönlein purpura) after COVID-19: A case series and review of the literature. Nefrologia. (2022) 42:481–9. doi: 10.1016/j.nefroe.2022.11.003

PubMed Abstract | Crossref Full Text | Google Scholar

33. Roan R, Lam J, Chang D. COVID-19 unmasking concurrent IgA nephropathy and minimal change disease: TH-PO738. J Am Soc Nephrol. (2023) 34:296.

Google Scholar

34. Jung J, Lee J, Lee J. Kidney involvement in children during the SARS-CoV-2 Omicron variant pandemic. BMC Pediatr. (2023) 23:491. doi: 10.1186/s12887-023-04322-5

PubMed Abstract | Crossref Full Text | Google Scholar

35. Qi Z, Yuan S, Wei J, Xia S, Huang Y, Chen X, et al. Clinical and pathological features of omicron variant of SARS-CoV-2-associated kidney injury. J Med Virol. (2023) 95:e29196. doi: 10.1002/jmv.29196

PubMed Abstract | Crossref Full Text | Google Scholar

36. de Las Mercedes Noriega M, Husain-Syed F, Wulf S, Csala B, Krebs C, Jabs W, et al. Kidney biopsy findings in patients with SARS-CoV-2 infection or after COVID-19 vaccination. Clin J Am Soc Nephrol. (2023) 18:613–25. doi: 10.2215/CJN.0000000000000106

PubMed Abstract | Crossref Full Text | Google Scholar

37. Papalia G, Barbuto S, Campus A, Vischini G. Double glomerulopathies or two-faced janus? A challenging case in the COVID-19 era. J Nephrol. (2023) 36:225–8. doi: 10.1007/s40620-022-01351-8

PubMed Abstract | Crossref Full Text | Google Scholar

38. Ozcan S, Sonmez O, Karaca C, Ozdede A, Seyahi N. ANCA-associated vasculitis flare might be provoked by COVID-19 infection: A case report and a review of the literature. Clin Kidney J. (2022) 15:1987–95. doi: 10.1093/ckj/sfac186

PubMed Abstract | Crossref Full Text | Google Scholar

39. Vivekanand N, Naresh Singh R, Kumari N, Ranjan R, Saini S. A Novel Association Between Coronavirus Disease 2019 and Normocomplementemic Rapidly Progressive Glomerulonephritis-Crescentic Immunoglobulin A Nephropathy: A Report of Two Pediatric Cases. (2022). Available online at: https://www.researchgate.net/publication/360874455_A_Novel_Association_Between_Coronavirus_Disease_2019_and_Normocomplementemic_Rapidly_Progressive_Glomerulonephritis-Crescentic_Immunoglobulin_A_Nephropathy_A_Report_of_Two_Pediatric_Cases (accessed September 25, 2024).

Google Scholar

40. Yonishi H, Katada R, Kofune K, Kusunoki Y, Ikeda N, Ueno N, et al. New-onset immunoglobulin-A nephropathy post severe acute respiratory syndrome-coronavirus-2 infection indicates rapidly progressive glomerulonephritis. Nephrology. (2022) 27:542–3. doi: 10.1111/nep.14003

PubMed Abstract | Crossref Full Text | Google Scholar

41. Shenoy S, Rao I, Prabhu R, Nagaraju S, Paramasivam G, Rangaswamy D, et al. Crescentic glomerulonephritis due to coexistent IgA nephropathy and anti-glomerular basement membrane disease in a patient with COVID-19 disease: A case report. Nephrology. (2022) 27:727–8. doi: 10.1111/nep.14076

PubMed Abstract | Crossref Full Text | Google Scholar

42. Ahlers C, Wang B, Howell D, Choksi V. Extensive palpable purpura preceding renal dysfunction in immunoglobulin a vasculitis due to Coronavirus-19 infection. Am J Med. (2023) 136:655–8. doi: 10.1016/j.amjmed.2023.03.028

PubMed Abstract | Crossref Full Text | Google Scholar

43. Nomura E, Finn L, Bauer A, Rozansky D, Iragorri S, Jenkins R, et al. Pathology findings in pediatric patients with COVID-19 and kidney dysfunction. Pediatr Nephrol. (2022) 37:2375–81. doi: 10.1007/s00467-022-05457-w

PubMed Abstract | Crossref Full Text | Google Scholar

44. Kudose S, Santoriello D, Bomback A, Sekulic M, Batal I, Stokes M, et al. Longitudinal outcomes of COVID-19-associated collapsing glomerulopathy and other podocytopathies. J Am Soc Nephrol. (2021) 32:2958–69. doi: 10.1681/ASN.2021070931

PubMed Abstract | Crossref Full Text | Google Scholar

45. Anderegg M, Liu M, Saganas C, Montani M, Vogt B, Huynh-Do U, et al. De novo vasculitis after mRNA-1273 (Moderna) vaccination. Kidney Int. (2021) 100:474–6. doi: 10.1016/j.kint.2021.05.016

PubMed Abstract | Crossref Full Text | Google Scholar

46. Abramson M, Mon-Wei Yu S, Campbell K, Chung M, Salem F. IgA Nephropathy After SARS-CoV-2 Vaccination. Kidney Med. (2021) 3:860–3. doi: 10.1016/j.xkme.2021.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

47. Fenoglio R, Lalloni S, Marchisio M, Oddone V, De Simone E, Del Vecchio G, et al. New onset biopsy-proven nephropathies after COVID Vaccination. Am J Nephrol. (2022) 53:325–30. doi: 10.1159/000523962

PubMed Abstract | Crossref Full Text | Google Scholar

48. Caza T, Cassol C, Messias N, Hannoudi A, Haun R, Walker P, et al. Glomerular disease in temporal association with SARS-CoV-2 vaccination: A series of 29 Cases. Kidney360. (2021) 2:1770–80. doi: 10.34067/KID.0005372021

PubMed Abstract | Crossref Full Text | Google Scholar

49. Morimoto N, Mori T, Shioji S, Taguchi T, Watanabe H, Sakai K, et al. Rapidly progressive IgA nephropathy with membranoproliferative glomerulonephritis-like lesions in an elderly man following the third dose of an mRNA COVID-19 vaccine: A case report. BMC Nephrol. (2023) 24:108. doi: 10.1186/s12882-023-03169-3

PubMed Abstract | Crossref Full Text | Google Scholar

50. Qu L, Zhang A, Xu G, Wang X, Lou H, Lou J, et al. Successful treatment of new-onset diabetes mellitus and IgA nephropathy after COVID-19 vaccination: A case report. Am J Transl Res. (2023) 15:6626–31.

Google Scholar

51. Mima A, Lee S. IgA nephropathy after COVID-19 vaccination and analysis of reported cases. Heliyon. (2023) 9:e17206. doi: 10.1016/j.heliyon.2023.e17206

PubMed Abstract | Crossref Full Text | Google Scholar

52. Zhang Y, Liu X, Zhang Z, Zhou T, Zhang X, Yang H, et al. Clinicopathological and immunological features of new onset kidney disease: A rare event after SARS-CoV-2 vaccination. Natl Sci Rev. (2023) 10:nwac034. doi: 10.1093/nsr/nwac034

PubMed Abstract | Crossref Full Text | Google Scholar

53. Chen C, Yang S, Hsu Y, Sung C, Chu P, Wu C, et al. Acute kidney disease following COVID-19 vaccination: A single-center retrospective study. Front Med. (2023) 10:1189243. doi: 10.3389/fmed.2023.1189243

PubMed Abstract | Crossref Full Text | Google Scholar

54. Martínez-Ortega J, Perez-Hernandez F, Fernández-Reyna I, Eljure Lopez N. First onset of IgA vasculitis and nephritis following COVID-19 vaccination. Cureus. (2023) 15:e42448. doi: 10.7759/cureus.42448

PubMed Abstract | Crossref Full Text | Google Scholar

55. Waldman M, Sinaii N, Lerma E, Kurien A, Jhaveri K, Uppal N, et al. COVID-19 vaccination and new onset glomerular disease: Results from the IRocGN2 international registry. Kidney360. (2023) 4:349–62. doi: 10.34067/KID.0006832022

PubMed Abstract | Crossref Full Text | Google Scholar

56. Alzoabi N, Alqahtani J, Algamdi B, Alghamdi N, Ahmad A, Alkaltham G, et al. IgA Vasculitis following AstraZeneca/Oxford COVID-19, case report. Acta Biomed. (2023) 94:e2023074. doi: 10.23750/abm.v94i6.12959

PubMed Abstract | Crossref Full Text | Google Scholar

57. Ritter A, Helmchen B, Gaspert A, Bleisch J, Fritschi B, Buchkremer F, et al. Clinical spectrum of gross haematuria following SARS-CoV-2 vaccination with mRNA vaccines. Clin Kidney J. (2022) 15:961–73. doi: 10.1093/ckj/sfab284

PubMed Abstract | Crossref Full Text | Google Scholar

58. Choi Y, Lee C, Kim K, Yoo W. Sudden Onset of IgA vasculitis affecting vital organs in adult patients following SARS-CoV-2 Vaccines. Vaccines. (2022) 10:923. doi: 10.3390/vaccines10060923

PubMed Abstract | Crossref Full Text | Google Scholar

59. Mokos M, Bašić-Jukić N. IgA nephropathy following SARS-CoV-2 vaccination in a renal transplant recipient with a history of aristolochic acid nephropathy. Ther Apher Dial. (2022) 26:667–8. doi: 10.1111/1744-9987.13765

PubMed Abstract | Crossref Full Text | Google Scholar

60. Okada M, Kikuchi E, Nagasawa M, Oshiba A, Shimoda M. An adolescent girl diagnosed with IgA nephropathy following the first dose of the COVID-19 vaccine. CEN Case Rep. (2022) 11:376–9. doi: 10.1007/s13730-021-00679-7

PubMed Abstract | Crossref Full Text | Google Scholar

61. Ota K, Yonekura Y, Saigan M, Goto K, Nishi S. Comparison of renal histopathology in three patients with gross hematuria after SARS-CoV-2 vaccination. CEN Case Rep. (2023) 12:176–83. doi: 10.1007/s13730-022-00743-w

PubMed Abstract | Crossref Full Text | Google Scholar

62. Tseng P, Chuang S, Pan Y, Shih H, Chang C, Huang S. Gross hematuria and IgA nephropathy flare-up following the first dose of Moderna vaccine: A case report. Medicine. (2022) 101:e32524. doi: 10.1097/MD.0000000000032524

PubMed Abstract | Crossref Full Text | Google Scholar

63. Abdel-Qader D, Hazza Alkhatatbeh I, Hayajneh W, Annab H, Al Meslamani A, Elmusa R. IgA nephropathy in a pediatric patient after receiving the first dose of Pfizer-BioNTech COVID-19 vaccine. Vaccine. (2022) 40:2528–30. doi: 10.1016/j.vaccine.2022.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

64. Niel O, Florescu C. IgA nephropathy presenting as rapidly progressive glomerulonephritis following first dose of COVID-19 vaccine. Pediatr Nephrol. (2022) 37:461–2. doi: 10.1007/s00467-021-05351-x

PubMed Abstract | Crossref Full Text | Google Scholar

65. Chen Y, Yang C, Tsai C, Ang M, Chou S, Chiang W, et al. Newly-diagnosed immunoglobulin A nephropathy with increased plasma galactose-deficient-IgA1 antibody associated with mRNA COVID-19 vaccination: A case report. J Int Med Res. (2022) 50:3000605221129674. doi: 10.1177/03000605221129674

PubMed Abstract | Crossref Full Text | Google Scholar

66. Ran E, Wang M, Wang Y, Liu R, Yi Y, Liu Y. New-onset crescent IgA nephropathy following the CoronaVac vaccine: A case report. Medicine. (2022) 101:e30066. doi: 10.1097/MD.0000000000030066

PubMed Abstract | Crossref Full Text | Google Scholar

67. Nakatani S, Mori K, Morioka F, Hirata C, Tsuda A, Uedono H, et al. New-onset kidney biopsy-proven IgA vasculitis after receiving mRNA-1273 COVID-19 vaccine: Case report. CEN Case Rep. (2022) 11:358–62. doi: 10.1007/s13730-021-00677-9

PubMed Abstract | Crossref Full Text | Google Scholar

68. Sugita K, Kaneko S, Hisada R, Harano M, Anno E, Hagiwara S, et al. Development of IgA vasculitis with severe glomerulonephritis after COVID-19 vaccination: A case report and literature review. CEN Case Rep. (2022) 11:436–41. doi: 10.1007/s13730-022-00695-1

PubMed Abstract | Crossref Full Text | Google Scholar

69. Kudose S, Friedmann P, Albajrami O, D’Agati V. Histologic correlates of gross hematuria following Moderna COVID-19 vaccine in patients with IgA nephropathy. Kidney Int. (2021) 100:468–9. doi: 10.1016/j.kint.2021.06.011

PubMed Abstract | Crossref Full Text | Google Scholar

70. Perrin P, Bassand X, Benotmane I, Bouvier N. Gross hematuria following SARS-CoV-2 vaccination in patients with IgA nephropathy. Kidney Int. (2021) 100:466–8. doi: 10.1016/j.kint.2021.05.022

PubMed Abstract | Crossref Full Text | Google Scholar

71. Negrea L, Rovin B. Gross hematuria following vaccination for severe acute respiratory syndrome coronavirus 2 in 2 patients with IgA nephropathy. Kidney Int. (2021) 99:1487. doi: 10.1016/j.kint.2021.03.002

PubMed Abstract | Crossref Full Text | Google Scholar

72. Rahim S, Lin J, Wang J. A case of gross hematuria and IgA nephropathy flare-up following SARS-CoV-2 vaccination. Kidney Int. (2021) 100:238. doi: 10.1016/j.kint.2021.04.024

PubMed Abstract | Crossref Full Text | Google Scholar

73. Horino T. IgA nephropathy flare-up following SARS-CoV-2 vaccination. QJM. (2021) 114:735–6. doi: 10.1093/qjmed/hcab223

PubMed Abstract | Crossref Full Text | Google Scholar

74. Tan H, Tan R, Choo J, Lim C, Tan C, Loh A, et al. Is COVID-19 vaccination unmasking glomerulonephritis? Kidney Int. (2021) 100:469–71. doi: 10.1016/j.kint.2021.05.009

PubMed Abstract | Crossref Full Text | Google Scholar

75. Lim C, Choo J, Tan C. COVID-19 Vaccination in Immunoglobulin A Nephropathy. Am J Kidney Dis. (2021) 78:617. doi: 10.1053/j.ajkd.2021.07.001

PubMed Abstract | Crossref Full Text | Google Scholar

76. Fukuda M, Kaneko T, Kawai T, Ishii H, Shimizu A. Secondary immunoglobulin A nephropathy with gross hematuria leading to rapidly progressive glomerulonephritis following severe acute respiratory syndrome coronavirus 2 vaccination: A case report. BMC Nephrol. (2023) 24:232. doi: 10.1186/s12882-023-03287-y

PubMed Abstract | Crossref Full Text | Google Scholar

77. Nakao H, Koseki T, Kato K, Yamada S, Tsuboi N, Takahashi K, et al. COVID-19 mRNA vaccination is associated with IgA nephropathy: An analysis of the Japanese adverse drug event report database. J Pharm Pharm Sci. (2023) 26:11453. doi: 10.3389/jpps.2023.11453

PubMed Abstract | Crossref Full Text | Google Scholar

78. Marchisio M, Lalloni S, Morrone M, Rabajoli G, Careddu A, Fenoglio R, et al. #5747 COVID vaccination related glomerulopathies and interstitial nephritis. Nephrol Dial Transplant. (2023) 38:gfad063c_5747. doi: 10.1093/ndt/gfad063c_5747

Crossref Full Text | Google Scholar

79. Kanamori H. Gross hematuria can be an impact of severe acute respiratory syndrome coronavirus 2 vaccination on immunoglobulin A nephropathy: A case report. J Med Case Rep. (2022) 16:273. doi: 10.1186/s13256-022-03514-4

PubMed Abstract | Crossref Full Text | Google Scholar

80. Lo W, Chan K. Gross haematuria after mRNA COVID-19 vaccination in two patients with histological and clinical diagnosis of IgA nephropathy. Nephrology. (2022) 27:110–1. doi: 10.1111/nep.13992

PubMed Abstract | Crossref Full Text | Google Scholar

81. Fujita Y, Yoshida K, Ichikawa D, Shibagaki Y, Yazawa M. Abrupt worsening of occult IgA nephropathy after the first dose of SARS-CoV-2 vaccination. CEN Case Rep. (2022) 11:302–8. doi: 10.1007/s13730-021-00670-2

PubMed Abstract | Crossref Full Text | Google Scholar

82. Rimoldi C, Gilberti M, Gesualdo L, Sossai G, Sinico R, Giglio E, et al. #5722 new onset or recurrent glomerulonephritis following SARS-CoV2 vaccination: A multicenter experience. Nephrol Dial Transplant. (2023) 38:gfad063c_5722.

Google Scholar

83. Ota Y, Kuroki R, Iwata M, Taira H, Matsuo S, Kamijo M, et al. Association between COVID-19 vaccination and relapse of glomerulonephritis. Clin Exp Nephrol. (2023) 27:236–42. doi: 10.1007/s10157-022-02299-6

PubMed Abstract | Crossref Full Text | Google Scholar

84. Schaubschlager T, Rajora N, Diep S, Kirtek T, Cai Q, Hendricks A, et al. De novo or recurrent glomerulonephritis and acute tubulointerstitial nephritis after COVID-19 vaccination: A report of six cases from a single center. Clin Nephrol. (2022) 97:289–97. doi: 10.5414/CN110794

PubMed Abstract | Crossref Full Text | Google Scholar

85. Morisawa K, Honda M. Two patients presenting IgA nephropathy after COVID-19 vaccination during a follow-up for asymptomatic hematuria. Pediatr Nephrol. (2022) 37:1695–6. doi: 10.1007/s00467-022-05518-0

PubMed Abstract | Crossref Full Text | Google Scholar

86. Watanabe S, Zheng S, Rashidi A. IgA nephropathy relapse following COVID-19 vaccination treated with corticosteroid therapy: Case report. BMC Nephrol. (2022) 23:135. doi: 10.1186/s12882-022-02769-9

PubMed Abstract | Crossref Full Text | Google Scholar

87. Udagawa T, Motoyoshi Y. Macroscopic hematuria in two children with IgA nephropathy remission following Pfizer COVID-19 vaccination. Pediatr Nephrol. (2022) 37:1693–4. doi: 10.1007/s00467-022-05517-1

PubMed Abstract | Crossref Full Text | Google Scholar

88. Yokote S, Ueda H, Shimizu A, Okabe M, Yamamoto K, Tsuboi N, et al. IgA nephropathy with glomerular capillary IgA deposition following SARS-CoV-2 mRNA vaccination: A report of three cases. CEN Case Rep. (2022) 11:499–505. doi: 10.1007/s13730-022-00707-0

PubMed Abstract | Crossref Full Text | Google Scholar

89. Uchiyama Y, Fukasawa H, Ishino Y, Nakagami D, Kaneko M, Yasuda H, et al. Sibling cases of gross hematuria and newly diagnosed IgA nephropathy following SARS-CoV-2 vaccination. BMC Nephrol. (2022) 23:216. doi: 10.1186/s12882-022-02843-2

PubMed Abstract | Crossref Full Text | Google Scholar

90. Lai K. Pathogenesis of IgA nephropathy. Nat Rev Nephrol. (2012) 8:275–83. doi: 10.1038/nrneph.2012.58

PubMed Abstract | Crossref Full Text | Google Scholar

91. Rollino C, Vischini G, Coppo R. IgA nephropathy and infections. J Nephrol. (2016) 29:463–8. doi: 10.1007/s40620-016-0265-x

PubMed Abstract | Crossref Full Text | Google Scholar

92. Yu H, Sun B, Fang Z, Zhao J, Liu X, Li Y, et al. Distinct features of SARS-CoV-2-specific IgA response in COVID-19 patients. Eur Respir J. (2020) 56:2001526. doi: 10.1183/13993003.01526-2020

PubMed Abstract | Crossref Full Text | Google Scholar

93. Nihei Y, Kishi M, Suzuki H, Koizumi A, Yoshida M, Hamaguchi S, et al. IgA Nephropathy with Gross Hematuria Following COVID-19 mRNA Vaccination. Intern Med. (2022) 61:1033–7. doi: 10.2169/internalmedicine.8787-21

PubMed Abstract | Crossref Full Text | Google Scholar

94. Allen A, Willis F, Beattie T, Feehally J. Abnormal IgA glycosylation in Henoch-Schönlein purpura restricted to patients with clinical nephritis. Nephrol Dial Transplant. (1998) 13:930–4. doi: 10.1093/ndt/13.4.930

PubMed Abstract | Crossref Full Text | Google Scholar

95. Sebastian R, Arunachalam J, Rajendran M. Temporal Clustering of Antiglomerular Basement Membrane Disease in COVID-19 Pandemic: A Case Series. Int J Nephrol Renovasc Dis. (2021) 14:393–8. doi: 10.2147/IJNRD.S333894

PubMed Abstract | Crossref Full Text | Google Scholar

96. Kudose S, Batal I, Santoriello D, Xu K, Barasch J, Peleg Y, et al. Kidney biopsy findings in patients with COVID-19. J Am Soc Nephrol. (2020) 31:1959–68. doi: 10.1681/ASN.2020060802

PubMed Abstract | Crossref Full Text | Google Scholar

97. Nahhal S, Halawi A, Basma H, Jibai A, Ajami Z. Anti-glomerular basement membrane disease as a potential complication of COVID-19: A case report and review of literature. Cureus. (2020) 12:e12089. doi: 10.7759/cureus.12089

PubMed Abstract | Crossref Full Text | Google Scholar

98. Sacker A, Kung V, Andeen N. Anti-GBM nephritis with mesangial IgA deposits after SARS-CoV-2 mRNA vaccination. Kidney Int. (2021) 100:471–2. doi: 10.1016/j.kint.2021.06.006

PubMed Abstract | Crossref Full Text | Google Scholar

99. Gupta R, Ellis B. Concurrent antiglomerular basement membrane nephritis and antineutrophil cytoplasmic autoantibody-mediated glomerulonephritis after second dose of SARS-CoV-2 mRNA vaccination. Kidney Int Rep. (2022) 7:127–8. doi: 10.1016/j.ekir.2021.10.020

PubMed Abstract | Crossref Full Text | Google Scholar

100. Prema J, Muthukumaran A, Haridas N, Fernando E, Seshadri J, Kurien A. Two cases of double-positive antineutrophil cytoplasmic autoantibody and antiglomerular basement membrane disease after BBV152/Covaxin vaccination. Kidney Int Rep. (2021) 6:3090–1. doi: 10.1016/j.ekir.2021.10.004

PubMed Abstract | Crossref Full Text | Google Scholar

101. Ting J, Barbir E, McRae S, Schachter M, De Luca L, Riazy M, et al. Double-positive anti-glomerular basement membrane antibody and myeloperoxidase antineutrophil cytoplasmic autoantibody-associated glomerulonephritis Post COVID-19 mRNA vaccine: A case series of 4 patients. Can J Kidney Health Dis. (2023) 10:20543581231153217. doi: 10.1177/20543581231153217

PubMed Abstract | Crossref Full Text | Google Scholar

102. Hoi S, Ogawa M, Munemura C, Takata T, Isomoto H. Atypical anti-glomerular basement membrane nephritis after the first dose of the severe acute respiratory syndrome coronavirus 2 mRNA Vaccine. Yonago Acta Med. (2023) 66:300–5. doi: 10.33160/yam.2023.05.008

PubMed Abstract | Crossref Full Text | Google Scholar

103. Coorey C, Phua E, Chou A, Shen Y, Mather A. Anti-GBM disease after oxford-astraZeneca ChAdOx1 nCoV-19 Vaccination: A report of two cases. Case Rep Nephrol Dial. (2022) 12:234–7. doi: 10.1159/000525737

PubMed Abstract | Crossref Full Text | Google Scholar

104. Ahmed M, Mohamed S, Alhussein H, Eltazi I, Sibira R, Abdulhadi A. COVID-19 vaccine as a potential triggering factor for anti-glomerular basement membrane (GBM) Disease: A case report and literature review. Cureus. (2022) 14:e29075. doi: 10.7759/cureus.29075

PubMed Abstract | Crossref Full Text | Google Scholar

105. Koc N, Yildirim T, Saglam A, Arici M, Erdem Y. A patient with COVID-19 and anti-glomerular basement membrane disease. Nefrologia. (2021) 41:471–3. doi: 10.1016/j.nefroe.2021.08.008

PubMed Abstract | Crossref Full Text | Google Scholar

106. Prendecki M, Clarke C, Cairns T, Cook T, Roufosse C, Thomas D, et al. Anti-glomerular basement membrane disease during the COVID-19 pandemic. Kidney Int. (2020) 98:780–1. doi: 10.1016/j.kint.2020.06.009

PubMed Abstract | Crossref Full Text | Google Scholar

107. Raval P. “Goodpasture’s Syndrome.,” xPharm: The Comprehensive Pharmacology Reference. Amsterdam: Elsevier (2007). p. 1–4.

Google Scholar

108. Kluth D, Rees A. Anti-glomerular basement membrane disease. J Am Soc Nephrol. (1999) 10:2446–53. doi: 10.1681/ASN.V10112446

PubMed Abstract | Crossref Full Text | Google Scholar

109. Guo W, Tan P, Baikunje S. Membranous nephropathy in a patient with COVID-19 infection. J Nephrol. (2022) 35:351–5. doi: 10.1007/s40620-021-01165-0

PubMed Abstract | Crossref Full Text | Google Scholar

110. Miao J, Fidler M, Nasr S, Larsen C, Zoghby Z. Membranous nephropathy in a patient with coronavirus disease 2019 (COVID-19): A case report. Clin Nephrol Case Stud. (2019) 9:11–8. doi: 10.5414/CNCS110379

PubMed Abstract | Crossref Full Text | Google Scholar

111. Mateus C, Theias Manso R, Martins A, Branco P. Membranous nephropathy after a recent SARS-CoV-2 infection. BMJ Case Rep. (2023) 16:e252468. doi: 10.1136/bcr-2022-252468

PubMed Abstract | Crossref Full Text | Google Scholar

112. Gueguen L, Loheac C, Saidani N, Khatchatourian L. Membranous nephropathy following anti-COVID-19 mRNA vaccination. Kidney Int. (2021) 100:1140–1. doi: 10.1016/j.kint.2021.08.006

PubMed Abstract | Crossref Full Text | Google Scholar

113. Da Y, Goh G, Khatri P. A case of membranous nephropathy following Pfizer-BioNTech mRNA vaccination against COVID-19. Kidney Int. (2021) 100:938–9. doi: 10.1016/j.kint.2021.07.016

PubMed Abstract | Crossref Full Text | Google Scholar

114. Vongchaiudomchoke T, Cheunsuchon B, Noppakun K. Membranous nephropathy following SARS-CoV-2 mRNA-1273 vaccination. Med Clin. (2023) 161:504–5. doi: 10.1016/j.medcli.2023.06.044

PubMed Abstract | Crossref Full Text | Google Scholar

115. Chavarot N, Padden M, Amrouche L, Malard S, Scemla A, Sberro-Soussan R, et al. De novo posttransplant membranous nephropathy following BNT162b2 mRNA COVID-19 vaccine in a kidney transplant recipient. Am J Transplant. (2022) 22:3188–9. doi: 10.1111/ajt.17166

PubMed Abstract | Crossref Full Text | Google Scholar

116. Paxton L, McMahon L, Wong L. De novo PLA2R positive membranous nephropathy following BNT162b2 mRNA COVID-19 vaccine. Intern Med J. (2022) 52:2191–2. doi: 10.1111/imj.15915

PubMed Abstract | Crossref Full Text | Google Scholar

117. Rashid W, Mousa H, Khan J, Ijaz F, Ezell GDA. Case of membranous nephropathy hypothesized to be associated With COVID-19 vaccine. Cureus. (2022) 14:e24245. doi: 10.7759/cureus.24245

PubMed Abstract | Crossref Full Text | Google Scholar

118. Aydın M, Yıldız A, Oruç A, Sezen M, Dilek K, Güllülü M, et al. Relapse of primary membranous nephropathy after inactivated SARS-CoV-2 virus vaccination. Kidney Int. (2021) 100:464–5. doi: 10.1016/j.kint.2021.05.001

PubMed Abstract | Crossref Full Text | Google Scholar

119. Behiri S, Ghalli F. Relapse of membranous glomerulonephritis with crescentic transformation following Covid-19 Vaccination: A Case Report. J Clin Rev Case Rep. (2023) 8:145–8. doi: 10.33140/JCRC.08.07.03

Crossref Full Text | Google Scholar

120. Larsen C, Messias N, Silva F, Messias E, Walker P. Determination of primary versus secondary membranous glomerulopathy utilizing phospholipase A2 receptor staining in renal biopsies. Mod Pathol. (2013) 26:709–15. doi: 10.1038/modpathol.2012.207

PubMed Abstract | Crossref Full Text | Google Scholar

121. Caza T, Hassen S, Dvanajscak Z, Kuperman M, Edmondson R, Herzog C, et al. NELL1 is a target antigen in malignancy-associated membranous nephropathy. Kidney Int. (2021) 99:967–76. doi: 10.1016/j.kint.2020.07.039

PubMed Abstract | Crossref Full Text | Google Scholar

122. Bomback A, Fervenza F. Membranous nephropathy: Approaches to treatment. Am J Nephrol. (2018) 47(Suppl 1):30–42. doi: 10.1159/000481635

PubMed Abstract | Crossref Full Text | Google Scholar

123. Beck L, Bonegio R, Lambeau G, Beck D, Powell D, Cummins T, et al. M-type phospholipase A2 receptor as target antigen in idiopathic membranous nephropathy. N Engl J Med. (2009) 361:11–21. doi: 10.1056/NEJMoa0810457

PubMed Abstract | Crossref Full Text | Google Scholar

124. Ağbaş A, Akkoç G, Kızılırmak C, Çalışkan Dolu N, Bayramoğlu E, Elevli M. Kidney Involvement in Pediatric COVID-19 Cases: A single-center experience. Turk Arch Pediatr. (2022) 57:558–62. doi: 10.5152/TurkArchPediatr.2022.22012

PubMed Abstract | Crossref Full Text | Google Scholar

125. Gambella A, Barreca A, Biancone L, Roccatello D, Peruzzi L, Besso L, et al. Spectrum of Kidney injury following COVID-19 Disease: Renal biopsy findings in a single Italian Pathology Service. Biomolecules. (2022) 12:298. doi: 10.3390/biom12020298

PubMed Abstract | Crossref Full Text | Google Scholar

126. Daneshgar N, Liang P, Michels C, Nester C, Harshman L, Dai D. Case report: Clinical and pathological findings of a recurrent C3 glomerulopathy with superimposed membranoproliferative glomerulonephritis pattern and cryoglobulinemia associated with COVID-19. Front Pediatr. (2022) 10:827466. doi: 10.3389/fped.2022.827466

PubMed Abstract | Crossref Full Text | Google Scholar

127. Sethi S, Nester C, Smith R. Membranoproliferative glomerulonephritis and C3 glomerulopathy: Resolving the confusion. Kidney Int. (2012) 81:434–41. doi: 10.1038/ki.2011.399

PubMed Abstract | Crossref Full Text | Google Scholar

128. Smith R, Appel G, Blom A, Cook H, D’Agati V, Fakhouri F, et al. C3 glomerulopathy - understanding a rare complement-driven renal disease. Nat Rev Nephrol. (2019) 15:129–43. doi: 10.1038/s41581-018-0107-2

PubMed Abstract | Crossref Full Text | Google Scholar

129. Sethi S, D’Costa M, Hermann S, Nasr S, Fervenza F. Immune-complex glomerulonephritis after COVID-19 infection. Kidney Int Rep. (2021) 6:1170–3. doi: 10.1016/j.ekir.2021.02.002

PubMed Abstract | Crossref Full Text | Google Scholar

130. Göndör G, Ksiazek S, Regele H, Kronbichler A, Knechtelsdorfer M, Säemann M. Development of crescentic membranoproliferative glomerulonephritis after COVID-19 vaccination. Clin Kidney J. (2022) 15:2340–2. doi: 10.1093/ckj/sfac222

PubMed Abstract | Crossref Full Text | Google Scholar

131. Haas M, Kaul S, Eustace JA. HIV-associated immune complex glomerulonephritis with “lupus-like” features: A clinicopathologic study of 14 cases. Kidney Int. (2005) 67:1381–90. doi: 10.1111/j.1523-1755.2005.00215.x

PubMed Abstract | Crossref Full Text | Google Scholar

132. Bruggeman L, Nelson P. Controversies in the pathogenesis of HIV-associated renal diseases. Nat Rev Nephrol. (2009) 5:574–81. doi: 10.1038/nrneph.2009.139

PubMed Abstract | Crossref Full Text | Google Scholar

133. Nobakht E, Cohen S, Rosenberg A, Kimmel P. HIV-associated immune complex kidney disease. Nat Rev Nephrol. (2016) 12:291–300. doi: 10.1038/nrneph.2015.216

PubMed Abstract | Crossref Full Text | Google Scholar

134. Basiratnia M, Derakhshan D, Yeganeh B, Derakhshan A. Acute necrotizing glomerulonephritis associated with COVID-19 infection: Report of two pediatric cases. Pediatr Nephrol. (2021) 36:1019–23. doi: 10.1007/s00467-021-04944-w

PubMed Abstract | Crossref Full Text | Google Scholar

135. Kim S, Jung J, Cho H, Lee J, Go H, Lee J. A child with crescentic glomerulonephritis following SARS-CoV-2 mRNA (Pfizer-BioNTech) vaccination. Pediatr Nephrol. (2023) 38:299–302. doi: 10.1007/s00467-022-05681-4

PubMed Abstract | Crossref Full Text | Google Scholar

136. Gupta R. Pauci-immune crescentic glomerulonephritis. Indian J Pathol Microbiol. (2003) 46:357–66.

Google Scholar

137. Bressler M, Pathak N, Cervellione K, Bagheri F, Epstein E, Mir A, et al. New onset granulomatosis with polyangiitis associated with COVID-19. Case Rep Dermatol Med. (2021) 2021:8877292. doi: 10.1155/2021/8877292

PubMed Abstract | Crossref Full Text | Google Scholar

138. Selvaraj V, Moustafa A, Dapaah-Afriyie K, Birkenbach M. COVID-19-induced granulomatosis with polyangiitis. BMJ Case Rep. (2021) 14:e242142. doi: 10.1136/bcr-2021-242142

PubMed Abstract | Crossref Full Text | Google Scholar

139. Purkayastha A, Reed G, Carino G, Birkenbach M. New onset granulomatosis with polyangiitis presenting as pulmonary-renal syndrome following covid-19 infection. Chest (2023) 164:A6225–6. doi: 10.1016/j.chest.2023.07.4005

Crossref Full Text | Google Scholar

140. Masiak A, Nowakowski S, Zdrojewski Z. SARS-CoV-2 infection associated with active granulomatosis with polyangiitis. Pol Arch Intern Med. (2021) 131:563–5. doi: 10.20452/pamw.15993

PubMed Abstract | Crossref Full Text | Google Scholar

141. Kaynar K, Güvercin B, Demir S, Mungan S, Çifçi E. A case report of antineutrophil cytoplasmic antibody-associated vasculitis and glomerular immune depositions. Hippokratia. (2022) 26:86–8.

Google Scholar

142. Mahdi S, Joudeh A, Raman K, Faqih S, Alhatou M, Wadiwala M, et al. New-onset severe eosinophilic granulomatosis with polyangiitis following the third dose of mRNA COVID-19 vaccine: A case report. Mod Rheumatol Case Rep. (2023) 8:153–8. doi: 10.1093/mrcr/rxad043

PubMed Abstract | Crossref Full Text | Google Scholar

143. Essien F, Evans J, Kyle A, Urisman A, Adams N. ’Granulomatosis with polyangiitis after Pfizer vaccination’: A case report. Ther Adv Rare Dis. (2022) 3:26330040221130084. doi: 10.1177/26330040221130084

PubMed Abstract | Crossref Full Text | Google Scholar

144. Davidovic T, Schimpf J, Sprenger-Mähr H, Abbassi-Nik A, Soleiman A, Zitt E, et al. De Novo and relapsing glomerulonephritis following SARS-CoV-2 mRNA vaccination in microscopic polyangiitis. Case Repo Nephrol. (2021) 2021:1–5. doi: 10.1155/2021/8400842

Crossref Full Text | Google Scholar

145. Avalos C, Ahmadzadeh Y, Gatsak D, Moosa S, Mozaffari M, Imas A, et al. Cardiac tamponade as a complication of microscopic polyangiitis: A case associated with a COVID-19 mRNA Vaccine. Cureus. (2023) 15:e37569. doi: 10.7759/cureus.37569

PubMed Abstract | Crossref Full Text | Google Scholar

146. Baier E, Olgemöller U, Biggemann L, Buck C, Tampe B. Dual-Positive MPO- and PR3-ANCA-associated vasculitis following SARS-CoV-2 mRNA booster vaccination: A case report and systematic review. Vaccines. (2022) 10:653. doi: 10.3390/vaccines10050653

PubMed Abstract | Crossref Full Text | Google Scholar

147. Okuda S, Hirooka Y, Sugiyama M. Propylthiouracil-induced antineutrophil cytoplasmic antibody-associated vasculitis after COVID-19 Vaccination. Vaccines. (2021) 9:842. doi: 10.3390/vaccines9080842

PubMed Abstract | Crossref Full Text | Google Scholar

148. Villa M, Díaz-Crespo F, Pérez de José A, Verdalles Ú, Verde E, Almeida Ruiz F, et al. A case of ANCA-associated vasculitis after AZD1222 (Oxford-AstraZeneca) SARS-CoV-2 vaccination: Casualty or causality? Kidney Int. (2021) 100:937–8. doi: 10.1016/j.kint.2021.07.026

PubMed Abstract | Crossref Full Text | Google Scholar

149. Obata S, Hidaka S, Yamano M, Yanai M, Ishioka K, Kobayashi S. MPO-ANCA-associated vasculitis after the Pfizer/BioNTech SARS-CoV-2 vaccination. Clin Kidney J. (2022) 15:357–9. doi: 10.1093/ckj/sfab181

PubMed Abstract | Crossref Full Text | Google Scholar

150. Chen C, Chen H, Lu C, Lin S. Case Report: Anti-neutrophil cytoplasmic antibody-associated vasculitis with acute renal failure and pulmonary hemorrhage may occur after COVID-19 vaccination. Front Med. (2021) 8:765447. doi: 10.3389/fmed.2021.765447

PubMed Abstract | Crossref Full Text | Google Scholar

151. Prabhahar A, Naidu G, Chauhan P, Sekar A, Sharma A, Sharma A, et al. ANCA-associated vasculitis following ChAdOx1 nCoV19 vaccination: Case-based review. Rheumatol Int. (2022) 42:749–58. doi: 10.1007/s00296-021-05069-x

PubMed Abstract | Crossref Full Text | Google Scholar

152. Gillion V, Jadoul M, Demoulin N, Aydin S, Devresse A. Granulomatous vasculitis after the AstraZeneca anti-SARS-CoV-2 vaccine. Kidney Int. (2021) 100:706–7. doi: 10.1016/j.kint.2021.06.033

PubMed Abstract | Crossref Full Text | Google Scholar

153. Tahir A, Walia J, Daly T, Gradzka A, Banai R. Rapidly progressive glomerulonephritis: A COVID-19 case report. Cureus. (2023) 15:e37767. doi: 10.7759/cureus.37767

PubMed Abstract | Crossref Full Text | Google Scholar

154. Thammathiwat T, Banjongjit A, Iampenkhae K, Townamchai N, Kanjanabuch T. ANCA associated glomerulonephritis following SARS-CoV-2 vaccination: A case series and systematic review. Vaccines. (2023) 11:983. doi: 10.3390/vaccines11050983

PubMed Abstract | Crossref Full Text | Google Scholar

155. Kitamoto K, Tanaka Y, Kuboyama T, Fujiki Y, Tomida K, Kamimori T, et al. Newly diagnosed ANCA-associated vasculitis after COVID-19 infection: A case report. J Med Case Rep. (2023) 17:366. doi: 10.1186/s13256-023-04081-y

PubMed Abstract | Crossref Full Text | Google Scholar

156. Chandok T, Nasr R, Uday K. A case of antineutrophil cytoplasmic antibody vasculitis-associated acute kidney injury in a patient with asymptomatic COVID-19 Infection. Cureus. (2023) 15:e35006. doi: 10.7759/cureus.35006

PubMed Abstract | Crossref Full Text | Google Scholar

157. Wnuk B, Heleniak Z, Buııo-Piontecka B, Liberek T, Dȩbska-Şlizień A. COVID-19 and vasculitis case report and review of the literature. Renal Dis Transpl Forum. (2023) 16:108–11. doi: 10.5603/rdtf.93319

Crossref Full Text | Google Scholar

158. Limaye H, Pathak J, Pathak H. A Case of De Novo antineutrophil cytoplasmic autoantibody associated glomerulonephritis following ChAdOx1 vaccination in an Indian Metropolitan City. Indian J Kidney Dis. (2023) 2:21.

Google Scholar

159. Sarwal A, Revelo P, Abraham J. #4600 MPO AND PR3 DUAL POSITIVE ANCA AND ANA: A CASE REPORT. Nephrol Dial Transplant. (2023) 38:gfad063c_4600.

Google Scholar

160. Mohamed M, Ibrahim A, Razaq Z, Hassan WA. Case of postcoronavirus disease 2019 antineutrophil cytoplasmic antibody-associated vasculitis successfully treated with rituximab. Saudi J Kidney Dis Transpl. (2019) 34(Suppl 1):S219–25. doi: 10.4103/sjkdt.sjkdt_317_22

PubMed Abstract | Crossref Full Text | Google Scholar

161. Cho W, Hwang S, Choi S, Kim B, Lee J, Lee D, et al. Multi-organ relapse following COVID-19 in myeloperoxidase-anti-neutrophil cytoplasmic antibody-associated vasculitis: A case report. Case Rep Nephrol Dial. (2023) 13:173–83. doi: 10.1159/000534331

PubMed Abstract | Crossref Full Text | Google Scholar

162. Rihem D, Azzabi A, Boukadida R, Mrabet S, Guedri Y, Wissal S, et al. #5034 ANCA associated vasculitis after COVID-19. Nephrol Dial Transplant. (2023) 38:gfad063d_5034. doi: 10.1093/ndt/gfad063d_5034

Crossref Full Text | Google Scholar

163. Ural Z, Yıldırım S, Şahin O, Tomar V, Zelyurt N, Koç H, et al. Successful treatment of post-COVID-19 severe ANCA-associated vasculitis: Case report. Res Square. (2023): doi: 10.21203/rs.3.rs-2802716/v1

Crossref Full Text | Google Scholar

164. Martín Navarro J, Cintra Cabrera M, Proccacini F, Muñoz Rodríguez J, Roldán Cortés D, Lucena Valverde R, et al. More difficult still: Treating severe rapidly progressive glomerulonephritis in the context of COVID-19 pneumonia. Nefrologia. (2022) 42:94–8. doi: 10.1016/j.nefroe.2020.12.007

PubMed Abstract | Crossref Full Text | Google Scholar

165. Lee J, Hwang H. Acquired antineutrophil cytoplasmic antibody-associated glomerulonephritis after COVID-19 infection. Kidney Res Clin Pract. (2023) 42:405–8. doi: 10.23876/j.krcp.22.261

PubMed Abstract | Crossref Full Text | Google Scholar

166. Latief M, Mir T, Wani M. Management of antineutrophil cytoplasmic antibody-associated vasculitis with COVID-19: A single center experience. Indian J Nephrol. (2023) 33:50–3. doi: 10.4103/ijn.ijn_423_21

PubMed Abstract | Crossref Full Text | Google Scholar

167. Schioppo T, Argolini L, Sciascia S, Pregnolato F, Tamborini F, Miraglia P, et al. Clinical and peculiar immunological manifestations of SARS-CoV-2 infection in systemic lupus erythematosus patients. Rheumatology. (2022) 61:1928–35. doi: 10.1093/rheumatology/keab611

PubMed Abstract | Crossref Full Text | Google Scholar

168. Babu S, Senthilnathan G, Shah S, Annigeri R. Double anti-neutrophil cytoplasmic antibody and anti-glomerular basement membrane antibody-positive crescentic glomerulonephritis, following SARS-CoV-2 Infection. Indian J Nephrol. (2022) 32:491–4. doi: 10.4103/ijn.ijn_344_21

PubMed Abstract | Crossref Full Text | Google Scholar

169. Briones E, Méndez G, Laguna J, Vargas M. [Association of COVID-19 and glomerulonephritis with ANCA and anti-glomerular basement membrane antibodies. Report of one case]. Rev Med Chil. (2022) 150:828–31. doi: 10.4067/S0034-98872022000600828

PubMed Abstract | Crossref Full Text | Google Scholar

170. Ta H, Awada H, Kang P, Gilbert N, Haller N, Mostow E, et al. Antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis with mucosal involvement following COVID-19 Pneumonia. Cureus. (2022) 14:e31441. doi: 10.7759/cureus.31441

PubMed Abstract | Crossref Full Text | Google Scholar

171. Uddin K, Mohamed K, Agboola A, Naqvi W, Hussaini H, Mohamed A, et al. Antineutrophil cytoplasmic antibody (ANCA)-associated renal vasculitis following COVID-19 vaccination: A case report and literature review. Cureus. (2022) 14:e30206. doi: 10.7759/cureus.30206

PubMed Abstract | Crossref Full Text | Google Scholar

172. Zakrocka I, Korolczuk A, Zaıuska W. Antineutrophil cytoplasmic autoantibody-associated vasculitis with rapid progressive glomerulonephritis following SARS-CoV-2 infection: A cause or coincidence? Pol Arch Intern Med. (2022) 132:16165. doi: 10.20452/pamw.16165

PubMed Abstract | Crossref Full Text | Google Scholar

173. Bansal S, Rana A, Manhas N, Rana A. Post COVID Vaccination (COVAXIN™ -BB152 V) Pauci-immune crescentic glomerulonephritis. Indian J Nephrol. (2022) 32:495–7. doi: 10.4103/ijn.ijn_352_21

PubMed Abstract | Crossref Full Text | Google Scholar

174. Kataria S, Rogers S, Sadia H, Ali T, Qureshi H, Bano S, et al. Antineutrophil cytoplasmic antibody (ANCA)-associated renal vasculitis after COVID-19 infection: A case report. Cureus. (2022) 14:e26111. doi: 10.7759/cureus.26111

PubMed Abstract | Crossref Full Text | Google Scholar

175. Yadav R, Shah S, Chhetri S. ANCA-associated vasculitis following Johnson and Johnson COVID-19 vaccine. Ann Med Surg. (2022) 79:104123. doi: 10.1016/j.amsu.2022.104123

PubMed Abstract | Crossref Full Text | Google Scholar

176. Kawamura T, Nakazawa D, Nishio S, Isozaki T, Komatsumoto M, Atsumi T. Development of ANCA-associated vasculitis followed by SARS-CoV-2 vaccination in a patient with HLA-DRB1*09:01 allele. Mod Rheumatol Case Rep. (2023) 7:426–30. doi: 10.1093/mrcr/rxac093

PubMed Abstract | Crossref Full Text | Google Scholar

177. Ural Z, Yıldırım S, Şahin O, Tomar V, Zelyurt N, Koç H, et al. COVID-19 infection can be associated with Pauci-Immune type of crescentic glomerulonephritis: A case report. Am J Clin Pathol. (2023) 160:S105–6. doi: 10.1093/ajcp/aqad150.232

PubMed Abstract | Crossref Full Text | Google Scholar

178. Uppal N, Kello N, Shah H, Khanin Y, De Oleo I, Epstein E, et al. De Novo ANCA-associated vasculitis with glomerulonephritis in COVID-19. Kidney Int Rep. (2020) 5:2079–83. doi: 10.1016/j.ekir.2020.08.012

PubMed Abstract | Crossref Full Text | Google Scholar

179. Moeinzadeh F, Dezfouli M, Naimi A, Shahidi S, Moradi H. Newly diagnosed glomerulonephritis during COVID-19 infection undergoing immunosuppression therapy, a case report. Iran J Kidney Dis. (2020) 14:239–42.

Google Scholar

180. Izci Duran T, Turkmen E, Dilek M, Sayarlioglu H, Arik N. ANCA-associated vasculitis after COVID-19. Rheumatol Int. (2021) 41:1523–9. doi: 10.1007/s00296-021-04914-3

PubMed Abstract | Crossref Full Text | Google Scholar

181. Hong Y, Shi P, Liu X, Yang L, Li K, Xu F, et al. Distinction between MPO-ANCA and PR3-ANCA-associated glomerulonephritis in Chinese patients: A retrospective single-center study. Clin Rheumatol. (2019) 38:1665–73. doi: 10.1007/s10067-019-04458-9

PubMed Abstract | Crossref Full Text | Google Scholar

182. Santoriello D, Bomback A, Kudose S, Batal I, Stokes M, Canetta P, et al. Anti-neutrophil cytoplasmic antibody associated glomerulonephritis complicating treatment with hydralazine. Kidney Int. (2021) 100:440–6. doi: 10.1016/j.kint.2021.03.029

PubMed Abstract | Crossref Full Text | Google Scholar

183. Söderberg D, Segelmark M. Neutrophil extracellular traps in ANCA-associated vasculitis. Front Immunol. (2016) 7:256. doi: 10.3389/fimmu.2016.00256

PubMed Abstract | Crossref Full Text | Google Scholar

184. Nakazawa D, Masuda S, Tomaru U, Ishizu A. Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nat Rev Rheumatol. (2019) 15:91–101. doi: 10.1038/s41584-018-0145-y

PubMed Abstract | Crossref Full Text | Google Scholar

185. Barnes B, Adrover J, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford J, et al. Targeting potential drivers of COVID-19: Neutrophil extracellular traps. J Exp Med. (2020) 217:e20200652. doi: 10.1084/jem.20200652

PubMed Abstract | Crossref Full Text | Google Scholar

186. Radermecker C, Detrembleur N, Guiot J, Cavalier E, Henket M, d’Emal C, et al. Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19. J Exp Med. (2020) 217:e20201012. doi: 10.1084/jem.20201012

PubMed Abstract | Crossref Full Text | Google Scholar

187. Sharma P, Uppal N, Wanchoo R, Shah H, Yang Y, Parikh R, et al. COVID-19-associated kidney injury: A case series of kidney biopsy findings. J Am Soc Nephrol. (2020) 31:1948–58. doi: 10.1681/ASN.2020050699

PubMed Abstract | Crossref Full Text | Google Scholar

188. Gupta S, Kaplan M. The role of neutrophils and NETosis in autoimmune and renal diseases. Nat Rev Nephrol. (2016) 12:402–13. doi: 10.1038/nrneph.2016.71

PubMed Abstract | Crossref Full Text | Google Scholar

189. Sekar A, Campbell R, Tabbara J, Rastogi P. ANCA glomerulonephritis after the Moderna COVID-19 vaccination. Kidney Int. (2021) 100:473–4. doi: 10.1016/j.kint.2021.05.017

PubMed Abstract | Crossref Full Text | Google Scholar

190. Al-Yafeai Z, Horn B, Terraccaine W, Jose A, Krishnan P. A case of antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis post COVID-19 vaccination. Cureus. (2022) 14:e23162. doi: 10.7759/cureus.23162

PubMed Abstract | Crossref Full Text | Google Scholar

191. Feghali E, Zafar M, Abid S, Santoriello D, Mehta S. De-novo antineutrophil cytoplasmic antibody-associated vasculitis following the mRNA-1273 (Moderna) Vaccine for COVID-19. Cureus. (2021) 13:e19616. doi: 10.7759/cureus.19616

PubMed Abstract | Crossref Full Text | Google Scholar

192. Tonutti A, Simonetta E, Stainer A, Suigo G, De Santis M, Selmi C, et al. Hepatic and pulmonary involvement in a patient with PR3-ANCA vasculitis following SARS-CoV-2 vaccination: A case report. Mod Rheumatol Case Rep. (2023) 7:440–3. doi: 10.1093/mrcr/rxad005

PubMed Abstract | Crossref Full Text | Google Scholar

193. Jha S, Hema A, Padmanabha H, Mahadevan A, Mailankody P, Mathuranath P, et al. ANCA-associated vasculitic neuropathy following Anti-SARS-CoV-2 vaccination: An epiphenomenon or causal association? Ann Indian Acad Neurol. (2023) 26:187–9. doi: 10.4103/aian.aian_848_22

PubMed Abstract | Crossref Full Text | Google Scholar

194. Garcia D, Martins C, da Fonseca E, de Carvalho V, de Rezende R. Clinical images: Severe proteinase 3 antineutrophil cytoplasmic antibody glomerulonephritis temporally associated with Sinovac Biotech’s inactivated SARS-CoV-2 vaccine. ACR Open Rheumatol. (2022) 4:277–8. doi: 10.1002/acr2.11397

PubMed Abstract | Crossref Full Text | Google Scholar

195. Shakoor M, Birkenbach M, Lynch M. ANCA-associated vasculitis following pfizer-bioNTech COVID-19 Vaccine. Am J Kidney Dis. (2021) 78:611–3. doi: 10.1053/j.ajkd.2021.06.016

PubMed Abstract | Crossref Full Text | Google Scholar

196. Dube G, Benvenuto L, Batal I. Antineutrophil cytoplasmic autoantibody-associated glomerulonephritis following the Pfizer-BioNTech COVID-19 Vaccine. Kidney Int Rep. (2021) 6:3087–9. doi: 10.1016/j.ekir.2021.08.012

PubMed Abstract | Crossref Full Text | Google Scholar

197. Hakroush S, Tampe B. Case Report: Anca-associated vasculitis presenting with rhabdomyolysis and pauci-immune crescentic glomerulonephritis after Pfizer-BioNTech COVID-19 mRNA vaccination. Front Immunol. (2021) 12:762006. doi: 10.3389/fimmu.2021.762006

PubMed Abstract | Crossref Full Text | Google Scholar

198. El Hasbani G, Uthman I. ANCA-associated vasculitis following the first dose of Pfizer-BioNTech COVID-19 Vaccine. Nephron. (2023) 147:103–7. doi: 10.1159/000525562

PubMed Abstract | Crossref Full Text | Google Scholar

199. Gen S, Iwai T, Ohnari S, Nobe K, Ikeda N. ANCA-associated vasculitis after moderna COVID-19 vaccination. Case Rep Nephrol. (2023) 2023:4906876. doi: 10.1155/2023/4906876

PubMed Abstract | Crossref Full Text | Google Scholar

200. Yoshino Y, Ishida T. Anti-neutrophil cytoplasmic antibody-associated vasculitis with periaortitis that developed after mRNA COVID-19 Vaccination. Cureus. (2023) 15:e37480. doi: 10.7759/cureus.37480

PubMed Abstract | Crossref Full Text | Google Scholar

201. Mazza F, Cicciarelli A, Rubino F, Leopizzi M, Di Maio V, Cerbelli B, et al. [Case report: Mpo-anca associated vasculitis after Pfizer-BioNTech COVID-19 mRNA Vaccination]. G Ital Nefrol. (2023) 40:2023.

Google Scholar

202. Zamoner W, Scardini J, De Dio B, Marques A, Silva V, Garcia A, et al. ANCA-associated vasculitis following Oxford-AstraZeneca COVID-19 vaccine in Brazil: Is there a causal relationship? A case report. Front Med. (2022) 9:1003332. doi: 10.3389/fmed.2022.1003332

PubMed Abstract | Crossref Full Text | Google Scholar

203. Cano-Gámez T, Teco-Cortes J, Soto-Abraham M, Álvarez-Hernández E. SARS-CoV-2 vaccination as a trigger for perinuclear antineutrophil cytoplasmic antibodies (p-ANCA) associated with rapidly progressive glomerulonephritis. Cureus. (2022) 14:e29924. doi: 10.7759/cureus.29924

PubMed Abstract | Crossref Full Text | Google Scholar

204. Noel E, Rashid U, Rabbani R, Khan W, Benjamin Y, Lee I. Antineutrophil cytoplasmic autoantibody-associated glomerulonephritis as a possible side effect of COVID-19 vaccination. Cureus. (2022) 14:e30565. doi: 10.7759/cureus.30565

PubMed Abstract | Crossref Full Text | Google Scholar

205. Suzuki M, Sekiguchi Y, Sasaki M, Inaba S, Oyama S, Inoue Y, et al. Antineutrophil cytoplasmic antibody-associated vasculitis after COVID-19 vaccination with Pfizer-BioNTech. Intern Med. (2022) 61:2925–9. doi: 10.2169/internalmedicine.9807-22

PubMed Abstract | Crossref Full Text | Google Scholar

206. Ma Y, Huang T, Xu G. ANCA-associated vasculitis following the CoronaVac vaccination. Ther Adv Chronic Dis. (2022) 13:20406223221125708. doi: 10.1177/20406223221125708

PubMed Abstract | Crossref Full Text | Google Scholar

207. Kim B, Kim H, Han K, Han S, Jo H. A Case Report of MPO-ANCA-associated vasculitis following heterologous mRNA1273 COVID-19 booster vaccination. J Korean Med Sci. (2022) 37:e204. doi: 10.3346/jkms.2022.37.e204

PubMed Abstract | Crossref Full Text | Google Scholar

208. Christodoulou M, Iatridi F, Chalkidis G, Lioulios G, Nikolaidou C, Badis K, et al. ANCA-associated vasculitis may result as a complication to both SARS-CoV-2 infection and vaccination. Life. (2022) 12:1072. doi: 10.3390/life12071072

PubMed Abstract | Crossref Full Text | Google Scholar

209. Zhang C, Maruggi G, Shan H, Li J. Advances in mRNA vaccines for infectious diseases. Front Immunol. (2019) 10:594. doi: 10.3389/fimmu.2019.00594

PubMed Abstract | Crossref Full Text | Google Scholar

210. Zhao L, David M, Hyjek E, Chang A, Meehan S. M2 macrophage infiltrates in the early stages of ANCA-associated pauci-immune necrotizing GN. Clin J Am Soc Nephrol. (2015) 10:54–62. doi: 10.2215/CJN.03230314

PubMed Abstract | Crossref Full Text | Google Scholar

211. Brunini F, Page T, Gallieni M, Pusey C. The role of monocytes in ANCA-associated vasculitides. Autoimmun Rev. (2016) 15:1046–53. doi: 10.1016/j.autrev.2016.07.031

PubMed Abstract | Crossref Full Text | Google Scholar

212. Unanue E, Beller D, Lu C, Allen P. Antigen presentation: Comments on its regulation and mechanism. J Immunol. (1984) 132:1–5. doi: 10.4049/jimmunol.132.1.1

Crossref Full Text | Google Scholar

213. Iking-Konert C, Vogt S, Radsak M, Wagner C, Hänsch G, Andrassy K. Polymorphonuclear neutrophils in Wegener’s granulomatosis acquire characteristics of antigen presenting cells. Kidney Int. (2001) 60:2247–62. doi: 10.1046/j.1523-1755.2001.00068.x

PubMed Abstract | Crossref Full Text | Google Scholar

214. Spies C, Kip M, Lau A, Sander M, Breuer J, Meyerhoefer J, et al. Influence of vaccination and surgery on HLA-DR expression in patients with upper aerodigestive tract cancer. J Int Med Res. (2008) 36:296–307. doi: 10.1177/147323000803600212

PubMed Abstract | Crossref Full Text | Google Scholar

215. Mantovani Cardoso E, Hundal J, Feterman D, Magaldi J. Concomitant new diagnosis of systemic lupus erythematosus and COVID-19 with possible antiphospholipid syndrome. Just a coincidence? A case report and review of intertwining pathophysiology. Clin Rheumatol. (2020) 39:2811–5. doi: 10.1007/s10067-020-05310-1

PubMed Abstract | Crossref Full Text | Google Scholar

216. Gracia-Ramos A, Saavedra-Salinas M. Can the SARS-CoV-2 infection trigger systemic lupus erythematosus? A case-based review. Rheumatol Int. (2021) 41:799–809. doi: 10.1007/s00296-021-04794-7

PubMed Abstract | Crossref Full Text | Google Scholar

217. Slimani Y, Abbassi R, El Fatoiki F, Barrou L, Chiheb S. Systemic lupus erythematosus and varicella-like rash following COVID-19 in a previously healthy patient. J Med Virol. (2021) 93:1184–7. doi: 10.1002/jmv.26513

PubMed Abstract | Crossref Full Text | Google Scholar

218. Mok C, Chu C, Tse S. De novo lupus nephritis after SARS-CoV-2 infection. Lupus. (2023) 32:893–9. doi: 10.1177/09612033231175280

PubMed Abstract | Crossref Full Text | Google Scholar

219. Kazzi B, Fine D, Geetha D, Chung M, Monroy-Trujillo M, Timlin H. New-onset lupus nephritis associated with COVID-19 infection. Lupus. (2022) 31:1007–11. doi: 10.1177/09612033221098571

PubMed Abstract | Crossref Full Text | Google Scholar

220. Zamani B, Moeini Taba S, Shayestehpour M. Systemic lupus erythematosus manifestation following COVID-19: A case report. J Med Case Rep. (2021) 15:29. doi: 10.1186/s13256-020-02582-8

PubMed Abstract | Crossref Full Text | Google Scholar

221. Ali S, Almas T, Zaidi U, Ahmed F, Shaikh S, Shaikh F, et al. A novel case of lupus nephritis and mixed connective tissue disorder in a COVID-19 patient. Ann Med Surg. (2022) 78:103653. doi: 10.1016/j.amsu.2022.103653

PubMed Abstract | Crossref Full Text | Google Scholar

222. Masset C, Renaudin K, Kervella D, Chapelet A, Deltombe C, Ville S. Collapsing glomerulopathy in a patient with APOL1 intermediate-risk genotype triggered by lupus nephritis and SARS-CoV-2 infection: Lessons for the clinical nephrologist. J Nephrol. (2022) 35:347–50. doi: 10.1007/s40620-021-01144-5

PubMed Abstract | Crossref Full Text | Google Scholar

223. Yusuf A, Cheong X, Rozita M, Periyasamy P, Ruslinda M. A case of lupus nephritis flare-up in severe COVID-19 infection. Med J Malaysia. (2021) 76:757–61.

Google Scholar

224. Alharthy A, Faqihi F, Nasim N, Noor A, Akhtar S, Balshi A, et al. COVID-19 in a patient with a flare of systemic lupus erythematosus: A rare case-report. Respir Med Case Rep. (2020) 31:101252. doi: 10.1016/j.rmcr.2020.101252

PubMed Abstract | Crossref Full Text | Google Scholar

225. Gayathri C, Monica K, Lakshmi P, Mathini S, Kumar N, Ram, et al. Systemic lupus erythematosus nephritis and COVID-19 disease. Clin Rheumatol. (2023) 42:2335–40. doi: 10.1007/s10067-023-06634-4

PubMed Abstract | Crossref Full Text | Google Scholar

226. Daniel E, Sekulic M, Kudose S, Kubin C, Ye X, Shayan K, et al. Kidney allograft biopsy findings after COVID-19. Am J Transplant. (2021) 21:4032–42. doi: 10.1111/ajt.16804

PubMed Abstract | Crossref Full Text | Google Scholar

227. Kim H, Jung M, Lim B, Han S. New-onset class III lupus nephritis with multi-organ involvement after COVID-19 vaccination. Kidney Int. (2022) 101:826–8. doi: 10.1016/j.kint.2022.01.013

PubMed Abstract | Crossref Full Text | Google Scholar

228. Zavala-Miranda M, González-Ibarra S, Pérez-Arias A, Uribe-Uribe N, Mejia-Vilet J. New-onset systemic lupus erythematosus beginning as class V lupus nephritis after COVID-19 vaccination. Kidney Int. (2021) 100:1340–1. doi: 10.1016/j.kint.2021.09.009

PubMed Abstract | Crossref Full Text | Google Scholar

229. Tuschen K, Bräsen J, Schmitz J, Vischedyk M, Weidemann A. Relapse of class V lupus nephritis after vaccination with COVID-19 mRNA vaccine. Kidney Int. (2021) 100:941–4. doi: 10.1016/j.kint.2021.07.019

PubMed Abstract | Crossref Full Text | Google Scholar

230. Sekar A. Lupus nephritis flare post Moderna mRNA-1273 coronavirus vaccine. QJM. (2022) 114:882–3. doi: 10.1093/qjmed/hcab284

PubMed Abstract | Crossref Full Text | Google Scholar

231. Hassanzadeh S, Mubarak M, Sepahi M, Nasri H. Exacerbation of an undiagnosed pre-existing lupus nephritis following an inactivated COVID-19 vaccination. J Nephropharmacol. (2021) 11:e2–2. doi: 10.34172/npj.2022.02

Crossref Full Text | Google Scholar

232. Muñoz L, Bertoli A, Rigo D, Sanchez Freytes M. [Relapse of lupus nephritis in temporal association with anti SARS-CoV-2 vaccination]. Medicina. (2022) 82:971–3.

Google Scholar

233. Weening J, D’Agati V, Schwartz M, Seshan S, Alpers C, Appel G, et al. The classification of glomerulonephritis in systemic lupus erythematosus revisited. J Am Soc Nephrol. (2004) 15:241–50. doi: 10.1097/01.asn.0000108969.21691.5d

PubMed Abstract | Crossref Full Text | Google Scholar

234. Bomback A, Kudose S, D’Agati V. De novo and relapsing glomerular diseases after COVID-19 vaccination: What do we know so far? Am J Kidney Dis. (2021) 78:477–80. doi: 10.1053/j.ajkd.2021.06.004

PubMed Abstract | Crossref Full Text | Google Scholar

235. Gupta R, Bhargava R, Shaukat A, Albert E, Leggat J. Spectrum of podocytopathies in new-onset nephrotic syndrome following COVID-19 disease: A report of 2 cases. BMC Nephrol. (2020) 21:326. doi: 10.1186/s12882-020-01970-y

PubMed Abstract | Crossref Full Text | Google Scholar

236. Akl A, Fakeeh M. WCN23-0542 SARS-COV2 associated minimal change glomerulonephritis long-term effects. Kidney Int Rep. (2023) 8:S70–1. doi: 10.1016/j.ekir.2023.02.156

Crossref Full Text | Google Scholar

237. Pokharel A, Anderson J, Deebajah M, Blatt N, Reddy G, Garlapaty V, et al. Podocytopathies related to either COVID-19 infection or its vaccination, our experience and literature review. Ultrastruct Pathol. (2023) 47:373–81. doi: 10.1080/01913123.2023.2237565

PubMed Abstract | Crossref Full Text | Google Scholar

238. Lebedev L, Sapojnikov M, Wechsler A, Varadi-Levi R, Zamir D, Tobar A, et al. Minimal change disease following the Pfizer-BioNTech COVID-19 Vaccine. Am J Kidney Dis. (2021) 78:142–5. doi: 10.1053/j.ajkd.2021.03.010

PubMed Abstract | Crossref Full Text | Google Scholar

239. Baskaran K, Cohen A, Weerasinghe N, Vilayur E. Report of two cases of minimal change disease following vaccination for COVID -19. Nephrology. (2022) 27:111–2. doi: 10.1111/nep.13995

PubMed Abstract | Crossref Full Text | Google Scholar

240. Thappy S, Thalappil S, Abbarh S, Al-Mashdali A, Akhtar M, Alkadi M. Minimal change disease following the Moderna COVID-19 vaccine: First case report. BMC Nephrol. (2021) 22:376. doi: 10.1186/s12882-021-02583-9

PubMed Abstract | Crossref Full Text | Google Scholar

241. Holzworth A, Couchot P, Cruz-Knight W, Brucculeri M. Minimal change disease following the Moderna mRNA-1273 SARS-CoV-2 vaccine. Kidney Int. (2021) 100:463–4. doi: 10.1016/j.kint.2021.05.007

PubMed Abstract | Crossref Full Text | Google Scholar

242. D’Agati V, Kudose S, Bomback A, Adamidis A, Tartini A. Minimal change disease and acute kidney injury following the Pfizer-BioNTech COVID-19 vaccine. Kidney Int. (2021) 100:461–3. doi: 10.1016/j.kint.2021.04.035

PubMed Abstract | Crossref Full Text | Google Scholar

243. Maas R, Gianotten S, van der Meijden W. An additional case of minimal change disease following the Pfizer-BioNTech COVID-19 Vaccine. Am J Kidney Dis. (2021) 78:312. doi: 10.1053/j.ajkd.2021.05.003

PubMed Abstract | Crossref Full Text | Google Scholar

244. Salem F, Rein J, Yu S, Abramson M, Cravedi P, Chung M. Report of three cases of minimal change disease following the second dose of mRNA SARS-CoV-2 COVID-19 vaccine. Kidney Int Rep. (2021) 6:2523–4. doi: 10.1016/j.ekir.2021.07.017

PubMed Abstract | Crossref Full Text | Google Scholar

245. Weijers J, Alvarez C, Hermans M. Post-vaccinal minimal change disease. Kidney Int. (2021) 100:459–61. doi: 10.1016/j.kint.2021.06.004

PubMed Abstract | Crossref Full Text | Google Scholar

246. Hanna J, Ingram A, Shao T. Minimal change disease after first dose of Pfizer-BioNTech COVID-19 Vaccine: A case report and review of minimal change disease related to COVID-19 Vaccine. Can J Kidney Health Dis. (2021) 8:20543581211058271. doi: 10.1177/20543581211058271

PubMed Abstract | Crossref Full Text | Google Scholar

247. Leclerc S, Royal V, Lamarche C, Laurin L. Minimal change disease with severe acute kidney injury following the oxford-astraZeneca COVID-19 Vaccine: A Case Report. Am J Kidney Dis. (2021) 78:607–10. doi: 10.1053/j.ajkd.2021.06.008

PubMed Abstract | Crossref Full Text | Google Scholar

248. Kobayashi S, Fugo K, Yamazaki K, Terawaki H. Minimal change disease soon after Pfizer-BioNTech COVID-19 vaccination. Clin Kidney J. (2021) 14:2606–7. doi: 10.1093/ckj/sfab156

PubMed Abstract | Crossref Full Text | Google Scholar

249. Anupama Y, Patel R, Vankalakunti M. Nephrotic syndrome following ChAdOx1 nCoV-19 vaccine against SARScoV-2. Kidney Int Rep. (2021) 6:2248. doi: 10.1016/j.ekir.2021.06.024

PubMed Abstract | Crossref Full Text | Google Scholar

250. Lim J, Han M, Kim Y, Kim M, Jung H, Choi J, et al. New-onset nephrotic syndrome after Janssen COVID-19 Vaccination: A case report and literature review. J Korean Med Sci. (2021) 36:e218. doi: 10.3346/jkms.2021.36.e218

PubMed Abstract | Crossref Full Text | Google Scholar

251. Dirim A, Safak S, Andac B, Garayeva N, Demir E, Artan A, et al. Minimal change disease following vaccination with CoronaVac. Clin Kidney J. (2021) 14:2268–9. doi: 10.1093/ckj/sfab123

PubMed Abstract | Crossref Full Text | Google Scholar

252. Komaba H, Wada T, Fukagawa M. Relapse of minimal change disease following the Pfizer-BioNTech COVID-19 Vaccine. Am J Kidney Dis. (2021) 78:469–70. doi: 10.1053/j.ajkd.2021.05.006

PubMed Abstract | Crossref Full Text | Google Scholar

253. Kervella D, Jacquemont L, Chapelet-Debout A, Deltombe C, Ville S. Minimal change disease relapse following SARS-CoV-2 mRNA vaccine. Kidney Int. (2021) 100:457–8. doi: 10.1016/j.kint.2021.04.033

PubMed Abstract | Crossref Full Text | Google Scholar

254. Morlidge C, El-Kateb S, Jeevaratnam P, Thompson B. Relapse of minimal change disease following the AstraZeneca COVID-19 vaccine. Kidney Int. (2021) 100:459. doi: 10.1016/j.kint.2021.06.005

PubMed Abstract | Crossref Full Text | Google Scholar

255. Mancianti N, Guarnieri A, Tripodi S, Salvo D, Garosi G. Minimal change disease following vaccination for SARS-CoV-2. J Nephrol. (2021) 34:1039–40. doi: 10.1007/s40620-021-01091-1

PubMed Abstract | Crossref Full Text | Google Scholar

256. Kobayashi N, Fujisawa H, Kumagai J, Tanabe M. New-onset minimal change disease following the Moderna COVID-19 vaccine. BMJ Case Rep. (2023) 16:e255144. doi: 10.1136/bcr-2023-255144

PubMed Abstract | Crossref Full Text | Google Scholar

257. Pirzadeh A, Emami S, Zuckerman J, Nobakht N. Exacerbation of Minimal Change Disease Following mRNA COVID-19 Vaccination. Am J Case Rep. (2023) 24:e941621. doi: 10.12659/AJCR.941621

PubMed Abstract | Crossref Full Text | Google Scholar

258. Krishna A, Singh P, Mazumdar P, Sharma A. Minimal change disease (MCD) following vaccination with ChAdOx1 nCoV-19 vaccine in a young Indian male: A case report. J Family Med Prim Care. (2022) 11:6568–70. doi: 10.4103/jfmpc.jfmpc_1082_22

PubMed Abstract | Crossref Full Text | Google Scholar

259. Watts A, Keller K, Lerner G, Rosales I, Collins A, Sekulic M, et al. Discovery of autoantibodies targeting nephrin in minimal change disease supports a novel autoimmune etiology. J Am Soc Nephrol. (2022) 33:238–52. doi: 10.1681/ASN.2021060794

PubMed Abstract | Crossref Full Text | Google Scholar

260. Cara-Fuentes G, Clapp W, Johnson R, Garin E. Pathogenesis of proteinuria in idiopathic minimal change disease: Molecular mechanisms. Pediatr Nephrol. (2016) 31:2179–89. doi: 10.1007/s00467-016-3379-4

PubMed Abstract | Crossref Full Text | Google Scholar

261. Crane C, Ingulli E. Response to: Focal segmental glomerulosclerosis recurrence in a young adult with kidney transplant after mRNA COVID-19 vaccination. Pediatr Nephrol. (2022) 37:2219. doi: 10.1007/s00467-022-05601-6

PubMed Abstract | Crossref Full Text | Google Scholar

262. Crane C, Phebus E, Ingulli E. Immunologic response of mRNA SARS-CoV-2 vaccination in adolescent kidney transplant recipients. Pediatr Nephrol. (2022) 37:449–53. doi: 10.1007/s00467-021-05256-9

PubMed Abstract | Crossref Full Text | Google Scholar

263. Basic-Jukic N, Coric M, Bulimbasic S, Dika Z, Juric I, Furic-Cunko V, et al. Histopathologic findings on indication renal allograft biopsies after recovery from acute COVID-19. Clin Transplant. (2021) 35:e14486. doi: 10.1111/ctr.14486

PubMed Abstract | Crossref Full Text | Google Scholar

264. Thorburn C, Samarapungavan D, Kanaan H, Cohn S, Jabbar K, Li W, et al. Focal Segmental Glomerulosclerosis (FSGS) progressing to collapsing glomerulopathy in renal transplant recipients with and without COVID-19 infection. Transplant Proc. (2022) 54:1465–70. doi: 10.1016/j.transproceed.2022.02.010

PubMed Abstract | Crossref Full Text | Google Scholar

265. Alawad M, Subahi E, Al-Ani H, Taha N, Kamal I. A case of crescentic glomerulonephritis in a patient with COVID-19 infection: A case report and literature review. Medicine. (2022) 101:e28754. doi: 10.1097/MD.0000000000028754

PubMed Abstract | Crossref Full Text | Google Scholar

266. Jha V, Akal R, Sharma A, Mahapatra D. Post Covishield (ChAdOx1 nCoV-19) vaccination: New onset focal segmental glomerulosclerosis resistant to steroid and calcineurin inhibitor. Indian J Nephrol. (2022) 32:378–83. doi: 10.4103/ijn.ijn_23_22

PubMed Abstract | Crossref Full Text | Google Scholar

267. Shabaka A, Rovirosa-Bigot S, Márquez C, Alonso Riaño M, Fernández-Juárez G. Acute kidney failure and nephrotic syndrome secondary to COVID-19-associated focal segmental glomerulosclerosis. Nefrologia. (2022) 42:727–9. doi: 10.1016/j.nefroe.2020.10.003

PubMed Abstract | Crossref Full Text | Google Scholar

268. Afonso R, Calças Marques R, Borges H, Carias E, Domingos A, Cabrita A, et al. Tip lesion variant of focal and segmental glomerulosclerosis in a COVID-19 patient. Case Rep Nephrol Dial. (2022) 12:248–54. doi: 10.1159/000528029

PubMed Abstract | Crossref Full Text | Google Scholar

269. Rivero J, Merino-López M, Olmedo R, Garrido-Roldan R, Moguel B, Rojas G, et al. Association between postmortem kidney biopsy findings and acute kidney injury from patients with SARS-CoV-2 (COVID-19). Clin J Am Soc Nephrol. (2021) 16:685–93. doi: 10.2215/CJN.16281020

PubMed Abstract | Crossref Full Text | Google Scholar

270. Nowak P, Forycka J, Cegielska N, Harendarz K, Wa̧growska-Danilewicz M, Danilewicz M, et al. Glucocorticoids induce partial remission of focal segmental glomerulosclerosis but not interstitial nephritis in COVID-19 Acute Kidney Injury in an APOL1 low-risk genotype white patient. Am J Case Rep. (2021) 22:e933462. doi: 10.12659/AJCR.933462

PubMed Abstract | Crossref Full Text | Google Scholar

271. Lim C, Goh B. Case series De Novo Focal Segmental Glomerulosclerosis (FSGS) Post Covid 19 vaccination: Case series in a single centre in Malaysia. Malays J Med Health Sci. (2023) 19:2636–9346. doi: 10.47836/mjmhs.19.6.47

Crossref Full Text | Google Scholar

272. Horinouchi T, Ueda C, Kitakado H, Yoshikawa N, Nozu K. Steroid resistant nephrotic syndrome with collapsing focal segmental glomerulosclerosis in a 12-year-old Japanese female after SARS-CoV-2 vaccination. J Nephrol. (2023) 36:1435–8. doi: 10.1007/s40620-023-01577-0

PubMed Abstract | Crossref Full Text | Google Scholar

273. Kadariya K, Soji-Ayoade D, Isaac S, Paul S. Potential role of high-dose steroids in the treatment of COVID-19-associated nephropathy. Cureus. (2023) 15:e35372. doi: 10.7759/cureus.35372

PubMed Abstract | Crossref Full Text | Google Scholar

274. Fulchiero R, Amaral S. Focal segmental glomerulosclerosis recurrence in a young adult with kidney transplant after mRNA COVID-19 vaccination. Pediatr Nephrol. (2022) 37:2217. doi: 10.1007/s00467-022-05564-8

PubMed Abstract | Crossref Full Text | Google Scholar

275. Wu H, Larsen C, Hernandez-Arroyo C, Mohamed M, Caza T, Sharshir M, et al. AKI and collapsing glomerulopathy associated with COVID-19 and APOL 1 high-risk genotype. J Am Soc Nephrol. (2020) 31:1688–95. doi: 10.1681/ASN.2020050558

PubMed Abstract | Crossref Full Text | Google Scholar

276. Larsen C, Bourne T, Wilson J, Saqqa O, Sharshir M. Collapsing glomerulopathy in a patient with COVID-19. Kidney Int Rep. (2020) 5:935–9. doi: 10.1016/j.ekir.2020.04.002

PubMed Abstract | Crossref Full Text | Google Scholar

277. Magoon S, Bichu P, Malhotra V, Alhashimi F, Hu Y, Khanna S, et al. COVID-19-related glomerulopathy: A report of 2 cases of collapsing focal segmental glomerulosclerosis. Kidney Med. (2020) 2:488–92. doi: 10.1016/j.xkme.2020.05.004

PubMed Abstract | Crossref Full Text | Google Scholar

278. Sharma Y, Nasr S, Larsen C, Kemper A, Ormsby A, Williamson S. COVID-19-associated collapsing focal segmental glomerulosclerosis: A report of 2 cases. Kidney Med. (2020) 2:493–7. doi: 10.1016/j.xkme.2020.05.005

PubMed Abstract | Crossref Full Text | Google Scholar

279. Shetty A, Tawhari I, Safar-Boueri L, Seif N, Alahmadi A, Gargiulo R, et al. COVID-19-associated glomerular disease. J Am Soc Nephrol. (2021) 32:33–40. doi: 10.1681/ASN.2020060804

PubMed Abstract | Crossref Full Text | Google Scholar

280. Peleg Y, Kudose S, D’Agati V, Siddall E, Ahmad S, Nickolas T, et al. Acute kidney injury due to collapsing glomerulopathy following COVID-19 Infection. Kidney Int Rep. (2020) 5:940–5. doi: 10.1016/j.ekir.2020.04.017

PubMed Abstract | Crossref Full Text | Google Scholar

281. Kissling S, Rotman S, Gerber C, Halfon M, Lamoth F, Comte D, et al. Collapsing glomerulopathy in a COVID-19 patient. Kidney Int. (2020) 98:228–31. doi: 10.1016/j.kint.2020.04.006

PubMed Abstract | Crossref Full Text | Google Scholar

282. Santoriello D, Khairallah P, Bomback A, Xu K, Kudose S, Batal I, et al. Postmortem kidney pathology findings in patients with COVID-19. J Am Soc Nephrol. (2020) 31:2158–67. doi: 10.1681/ASN.2020050744

PubMed Abstract | Crossref Full Text | Google Scholar

283. Malik I, Ladiwala N, Chinta S, Khan M, Patel K. Severe acute respiratory syndrome coronavirus 2 induced focal segmental glomerulosclerosis. Cureus. (2020) 12:e10898. doi: 10.7759/cureus.10898

PubMed Abstract | Crossref Full Text | Google Scholar

284. Roy S, Kunaparaju S, Koduri N, Sangani V, Pokal M, Konala V, et al. COVID-19 and APOL-1 high-risk genotype-associated collapsing glomerulonephritis. Case Rep Nephrol. (2021) 2021:3737751. doi: 10.1155/2021/3737751

PubMed Abstract | Crossref Full Text | Google Scholar

285. Laboux T, Gibier J, Pottier N, Glowacki F, Hamroun A. COVID-19-related collapsing glomerulopathy revealing a rare risk variant of APOL1: Lessons for the clinical nephrologist. J Nephrol. (2021) 34:373–8. doi: 10.1007/s40620-020-00935-6

PubMed Abstract | Crossref Full Text | Google Scholar

286. Akilesh S, Nast C, Yamashita M, Henriksen K, Charu V, Troxell M, et al. Multicenter clinicopathologic correlation of kidney biopsies performed in COVID-19 patients presenting with acute kidney injury or proteinuria. Am J Kidney Dis. (2021) 77:82–93.e1. doi: 10.1053/j.ajkd.2020.10.001

PubMed Abstract | Crossref Full Text | Google Scholar

287. Tancredi T, DeWaters A, McGillen K. Renal ultrasound findings secondary to COVID-19 related collapsing focal segmental glomerulosclerosis - A case report. Clin Imaging. (2021) 71:34–8. doi: 10.1016/j.clinimag.2020.11.011

PubMed Abstract | Crossref Full Text | Google Scholar

288. Ferlicot S, Jamme M, Gaillard F, Oniszczuk J, Couturier A, May O, et al. The spectrum of kidney biopsies in hospitalized patients with COVID-19, acute kidney injury, and/or proteinuria. Nephrol Dial Transplant. (2021) [Online ahead of print]. doi: 10.1093/ndt/gfab042

PubMed Abstract | Crossref Full Text | Google Scholar

289. Kim A, Mahgoub A, Parajuli A, Towfiq B. COVID-19-associated nephropathy: A devastating complication. Cureus. (2023) 15:e43558. doi: 10.7759/cureus.43558

PubMed Abstract | Crossref Full Text | Google Scholar

290. Dyatlova N. ICollapsing glomerulopathy following moderna COVID-19 vaccine and response to treatment. Ann Clin Med Case Rep. (2023) 10:1–3.

Google Scholar

291. Malhotra V, Magoon S, Troyer D, McCune T. Collapsing focal segmental glomerulosclerosis and acute oxalate nephropathy in a patient with COVID-19: A double whammy. J Investig Med High Impact Case Rep. (2020) 8:2324709620963635. doi: 10.1177/2324709620963635

PubMed Abstract | Crossref Full Text | Google Scholar

292. Thomas A, Trainor R, Sheingold Z, Samarneh M. A case of COVID-19-associated focal segmental glomerulosclerosis. Cureus. (2023) 15:e37547. doi: 10.7759/cureus.37547

PubMed Abstract | Crossref Full Text | Google Scholar

293. Dhillon V, Alkashash A, Viquez-Beita K. Coronavirus disease 2019-associated nephropathy in an African American patient: A case report and review of the literature. J Med Case Rep. (2023) 17:153. doi: 10.1186/s13256-023-03888-z

PubMed Abstract | Crossref Full Text | Google Scholar

294. Kesiena O, Papadopoulos P, Amakye D, Hama E, Mackay R. COVID-19 associated collapsing glomerulopathy presenting as acute kidney injury on chronic kidney disease: A case report and review of the literature. CEN Case Rep. (2022) 11:273–7. doi: 10.1007/s13730-021-00667-x

PubMed Abstract | Crossref Full Text | Google Scholar

295. Neves P, Caires R, Guimarães M, Costalonga E, Cavalcante L, Costa E, et al. Collapsing glomerulopathy following SARS-CoV-2 adenovirus-vector-based vaccine: Report of 2 cases. Kidney Int. (2022) 101:637–9. doi: 10.1016/j.kint.2021.12.016

PubMed Abstract | Crossref Full Text | Google Scholar

296. Craig T, Ansari M, Foster P, Abdelgadir Y, Abdelghani A, Jha P. A Case of COVID-associated nephropathy (COVAN). Cureus. (2022) 14:e30872. doi: 10.7759/cureus.30872

PubMed Abstract | Crossref Full Text | Google Scholar

297. Maldonado D, Ray J, Lin X, Salem F, Brown M, Bansal I. COVAN leading to ESKD despite minimal COVID symptoms. J Investig Med High Impact Case Rep. (2022) 10:23247096221093888. doi: 10.1177/23247096221093888

PubMed Abstract | Crossref Full Text | Google Scholar

298. Alotaibi M, Ellis C, Wadhwani S, Peleg Y. A rare case of granulomatous interstitial nephritis in a patient with COVID-19. J Investig Med High Impact Case Rep. (2022) 10:23247096221114517. doi: 10.1177/23247096221114517

PubMed Abstract | Crossref Full Text | Google Scholar

299. Jefferis J, Kassianos A, Grivei A, Doucet B, Healy H, Francis L, et al. SARS-CoV-2 vaccination-associated collapsing glomerulopathy in a kidney transplant recipient. Kidney Int. (2022) 101:635–6. doi: 10.1016/j.kint.2021.12.018

PubMed Abstract | Crossref Full Text | Google Scholar

300. Nlandu Y, Makulo J, Pakasa N, Sumaili E, Nkondi C, Bukabau J, et al. First Case of COVID-19-associated collapsing glomerulopathy in Sub-Saharan Africa. Case Rep Nephrol. (2020) 2020:8820713. doi: 10.1155/2020/8820713

PubMed Abstract | Crossref Full Text | Google Scholar

301. Izzedine H, Brocheriou I, Arzouk N, Seilhean D, Couvert P, Cluzel P, et al. COVID-19-associated collapsing glomerulopathy: A report of two cases and literature review. Intern Med J. (2020) 50:1551–8. doi: 10.1111/imj.15041

PubMed Abstract | Crossref Full Text | Google Scholar

302. Giannini G, Carlos Q, Velez J, May R, Sharma S, Mohamed M, et al. Renal prognosis of COVID-19 associated nephropathy. Kidney Int Rep. (2022) 7:2722–5. doi: 10.1016/j.ekir.2022.09.027

PubMed Abstract | Crossref Full Text | Google Scholar

303. Hoilat G, Das G, Shahnawaz M, Shanley P, Bukhari S. COVID-19 induced collapsing glomerulopathy and role of APOL1. QJM. (2021) 114:263–4. doi: 10.1093/qjmed/hcaa335

PubMed Abstract | Crossref Full Text | Google Scholar

304. Velez J, Caza T, Larsen C. COVAN is the new HIVAN: The re-emergence of collapsing glomerulopathy with COVID-19. Nat Rev Nephrol. (2020) 16:565–7. doi: 10.1038/s41581-020-0332-3

PubMed Abstract | Crossref Full Text | Google Scholar

305. Wiggins B, Deliwala S, Banno F, Knight K, Minaudo M. Acute renal failure and nephrotic syndrome secondary to collapsing glomerulopathy associated with hepatitis C. Cureus. (2022) 14:e23175. doi: 10.7759/cureus.23175

PubMed Abstract | Crossref Full Text | Google Scholar

306. Gandhi P, Dowling C, Satoskar A, Shah A. Successful treatment of COVID-19-associated collapsing glomerulopathy: 22 months of follow-up. Clin Nephrol Case Stud. (2023) 11:110–3. doi: 10.5414/CNCS111112

PubMed Abstract | Crossref Full Text | Google Scholar

307. Rivera F, Ansay M, Golbin J, Alfonso P, Mangubat G, Menghrajani R, et al. HIV-associated nephropathy in 2022. Glomerular Dis. (2022) 3:1–11. doi: 10.1159/000526868

PubMed Abstract | Crossref Full Text | Google Scholar

308. Gaillard F, Ismael S, Sannier A, Tarhini H, Volpe T, Greze C, et al. Tubuloreticular inclusions in COVID-19-related collapsing glomerulopathy. Kidney Int. (2020) 98:241. doi: 10.1016/j.kint.2020.04.022

PubMed Abstract | Crossref Full Text | Google Scholar

309. Markowitz G, Nasr S, Stokes M, D’Agati V. Treatment with IFN-{alpha}, -{beta}, or -{gamma} is associated with collapsing focal segmental glomerulosclerosis. Clin J Am Soc Nephrol. (2010) 5:607–15. doi: 10.2215/CJN.07311009

PubMed Abstract | Crossref Full Text | Google Scholar

310. Rettmar K, Kienast J, van de Loo J. Minimal change glomerulonephritis with reversible proteinuria during interferon alpha 2a therapy for chronic myeloid leukemia. Am J Hematol. (1995) 49:355–6. doi: 10.1002/ajh.2830490417

PubMed Abstract | Crossref Full Text | Google Scholar

311. Zhou Z, Ren L, Zhang L, Zhong J, Xiao Y, Jia Z, et al. Heightened innate immune responses in the respiratory tract of COVID-19 Patients. Cell Host Microbe. (2020) 27:883–90.e2. doi: 10.1016/j.chom.2020.04.017

PubMed Abstract | Crossref Full Text | Google Scholar

312. Jamilloux Y, Henry T, Belot A, Viel S, Fauter M, El Jammal T, et al. Should we stimulate or suppress immune responses in COVID-19? Cytokine and anti-cytokine interventions. Autoimmun Rev. (2020) 19:102567. doi: 10.1016/j.autrev.2020.102567

PubMed Abstract | Crossref Full Text | Google Scholar

313. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. (2019) 395:497–506. doi: 10.1016/S0140-673630183-5

Crossref Full Text | Google Scholar

314. Batal I, Markowitz G, Wong W, Avasare R, Mapara M, Appel G, et al. Filgrastim-induced crescentic transformation of recurrent IgG2λ GN. J Am Soc Nephrol. (2016) 27:1911–5. doi: 10.1681/ASN.2016010061

PubMed Abstract | Crossref Full Text | Google Scholar

315. Halloran P, Venner J, Famulski K. Comprehensive analysis of transcript changes associated with allograft rejection: Combining universal and selective features. Am J Transplant. (2017) 17:1754–69. doi: 10.1111/ajt.14200

PubMed Abstract | Crossref Full Text | Google Scholar

316. Chen S, Kowalewska J, McCune T. COVID-19 associated collapsing FSGS in an APOL1 homozygous transplant recipient after successful COVID vaccination: A case report. Transplant Proc. (2022) 54:1543–6. doi: 10.1016/j.transproceed.2021.11.001

PubMed Abstract | Crossref Full Text | Google Scholar

317. Lazareth H, Péré H, Binois Y, Chabannes M, Schurder J, Bruneau T, et al. COVID-19-related collapsing glomerulopathy in a kidney transplant recipient. Am J Kidney Dis. (2020) 76:590–4. doi: 10.1053/j.ajkd.2020.06.009

PubMed Abstract | Crossref Full Text | Google Scholar

318. Kadosh B, Pavone J, Wu M, Reyentovich A, Gidea C. Collapsing glomerulopathy associated with COVID-19 infection in a heart transplant recipient. J Heart Lung Transplant. (2020) 39:855–7. doi: 10.1016/j.healun.2020.05.013

PubMed Abstract | Crossref Full Text | Google Scholar

319. Noble R, Tan M, McCulloch T, Shantier M, Byrne C, Hall M, et al. Collapsing glomerulopathy affecting native and transplant kidneys in individuals with COVID-19. Nephron. (2020) 144:589–94. doi: 10.1159/000509938

PubMed Abstract | Crossref Full Text | Google Scholar

320. Ribeiro B, Pontello Cristelli M, Demarchi Foresto R, Machado Proença H, Medina-Pestana J. Two-hit kidney allograft injury by SARS-CoV-2. Cureus. (2023) 15:e34603. doi: 10.7759/cureus.34603

PubMed Abstract | Crossref Full Text | Google Scholar

321. Levenson E, Shepherd T, Aviles D, Craver R, Ehlayel A, Love G, et al. De novo collapsing glomerulopathy in a pediatric kidney transplant recipient with COVID-19 infection. Pediatr Transplant. (2021) 25:e14013. doi: 10.1111/petr.14013

PubMed Abstract | Crossref Full Text | Google Scholar

322. Skorecki K, Lee J, Langefeld C, Rosset S, Tzur S, Wasser W, et al. A null variant in the apolipoprotein L3 gene is associated with non-diabetic nephropathy. Nephrol Dial Transplant. (2018) 33:323–30. doi: 10.1093/ndt/gfw451

PubMed Abstract | Crossref Full Text | Google Scholar

323. Freedman B, Limou S, Ma L, Kopp J. APOL1-associated nephropathy: A key contributor to racial disparities in CKD. Am J Kidney Dis. (2018) 72(5 Suppl 1):S8–16. doi: 10.1053/j.ajkd.2018.06.020

PubMed Abstract | Crossref Full Text | Google Scholar

324. Friedman D, Pollak M. APOL1 and kidney disease: From genetics to biology. Annu Rev Physiol. (2020) 82:323–42. doi: 10.1146/annurev-physiol-021119-034345

PubMed Abstract | Crossref Full Text | Google Scholar

325. Nichols B, Jog P, Lee J, Blackler D, Wilmot M, D’Agati V, et al. Innate immunity pathways regulate the nephropathy gene Apolipoprotein L1. Kidney Int. (2015) 87:332–42. doi: 10.1038/ki.2014.270

PubMed Abstract | Crossref Full Text | Google Scholar

326. Chang J, Husain S, Santoriello D, Stokes M, Miles C, Foster K, et al. Donor’s APOL1 risk genotype and “Second Hits” associated with de novo collapsing glomerulopathy in deceased donor kidney transplant recipients: A report of 5 cases. Am J Kidney Dis. (2019) 73:134–9. doi: 10.1053/j.ajkd.2018.05.008

PubMed Abstract | Crossref Full Text | Google Scholar

327. Meliambro K, Li X, Salem F, Yi Z, Sun Z, Chan L, et al. Molecular analysis of the kidney from a patient with COVID-19-associated collapsing glomerulopathy. Kidney Med. (2021) 3:653–8. doi: 10.1016/j.xkme.2021.02.012

PubMed Abstract | Crossref Full Text | Google Scholar

328. Couturier A, Ferlicot S, Chevalier K, Guillet M, Essig M, Jauréguiberry S, et al. Indirect effects of severe acute respiratory syndrome coronavirus 2 on the kidney in coronavirus disease patients. Clin Kidney J. (2020) 13:347–53. doi: 10.1093/ckj/sfaa088

PubMed Abstract | Crossref Full Text | Google Scholar

329. Pan X, Xu D, Zhang H, Zhou W, Wang L, Cui X. Identification of a potential mechanism of acute kidney injury during the COVID-19 outbreak: A study based on single-cell transcriptome analysis. Intensive Care Med. (2020) 46:1114–6. doi: 10.1007/s00134-020-06026-1

PubMed Abstract | Crossref Full Text | Google Scholar

330. Chen Q, Li J, Xiang Z, Lang Y, Guo G, Liu Z. Localization of cell receptor-related genes of SARS-CoV-2 in the kidney through single-cell transcriptome analysis. Kidney Dis. (2020) 6:258–70. doi: 10.1159/000508162

PubMed Abstract | Crossref Full Text | Google Scholar

331. Varga Z, Flammer A, Steiger P, Haberecker M, Andermatt R, Zinkernagel A, et al. Endothelial cell infection and endotheliitis in COVID-19. Lancet. (2020) 395:1417–8. doi: 10.1016/S0140-673630937-5

Crossref Full Text | Google Scholar

332. Herbert S, Stainier D. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat Rev Mol Cell Biol. (2011) 12:551–64. doi: 10.1038/nrm3176

PubMed Abstract | Crossref Full Text | Google Scholar

333. Diao B, Wang C, Wang R, Feng Z, Zhang J, Yang H, et al. Human kidney is a target for novel severe acute respiratory syndrome coronavirus 2 infection. Nat Commun. (2021) 12:2506. doi: 10.1038/s41467-021-22781-1

PubMed Abstract | Crossref Full Text | Google Scholar

334. Westhoff T, Seibert F, Bauer F, Stervbo U, Anft M, Doevelaar A, et al. Allograft infiltration and meningoencephalitis by SARS-CoV-2 in a pancreas-kidney transplant recipient. Am J Transplant. (2020) 20:3216–20. doi: 10.1111/ajt.16223

PubMed Abstract | Crossref Full Text | Google Scholar

335. Peng Y, Mentzer A, Liu G, Yao X, Yin Z, Dong D, et al. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat Immunol. (2020) 21:1336–45. doi: 10.1038/s41590-020-0782-6

PubMed Abstract | Crossref Full Text | Google Scholar

336. Rha M, Shin E. Activation or exhaustion of CD8+ T cells in patients with COVID-19. Cell Mol Immunol. (2021) 18:2325–33. doi: 10.1038/s41423-021-00750-4

PubMed Abstract | Crossref Full Text | Google Scholar

337. Pozzetto B, Legros V, Djebali S, Barateau V, Guibert N, Villard M, et al. Immunogenicity and efficacy of heterologous ChAdOx1-BNT162b2 vaccination. Nature. (2021) 600:701–6. doi: 10.1038/s41586-021-04120-y

PubMed Abstract | Crossref Full Text | Google Scholar

338. Cairoli E, Espinosa G. Autoimmune diseases and vaccines against COVID-19. Decision making in uncertain scenarios. Med Clin. (2021) 157:247–52.

Google Scholar

339. Tipping P, Holdsworth S. T cells in crescentic glomerulonephritis. J Am Soc Nephrol. (2006) 17:1253–63. doi: 10.1681/ASN.2005091013

PubMed Abstract | Crossref Full Text | Google Scholar

340. Chan O, Madaio M, Shlomchik M. B cells are required for lupus nephritis in the polygenic, Fas-intact MRL model of systemic autoimmunity. J Immunol. (1999) 163:3592–6.

Google Scholar

341. Kopp J, Anders H, Susztak K, Podestà M, Remuzzi G, Hildebrandt F, et al. Podocytopathies. Nat Rev Dis Primers. (2020) 6:68. doi: 10.1038/s41572-020-0196-7

PubMed Abstract | Crossref Full Text | Google Scholar

342. Murakami M, Kamimura D, Hirano T. Pleiotropy and specificity: Insights from the interleukin 6 family of cytokines. Immunity. (2019) 50:812–31. doi: 10.1016/j.immuni.2019.03.027

PubMed Abstract | Crossref Full Text | Google Scholar

343. Bastolla U, Chambers P, Abia D, Garcia-Bermejo M, Fresno M. Is Covid-19 severity associated with ACE2 degradation? Front Drug Discov. (2022) 1:789710. doi: 10.3389/fddsv.2021.789710

Crossref Full Text | Google Scholar

344. Su C, Wang L, Yoo D. Activation of NF-κB and induction of proinflammatory cytokine expressions mediated by ORF7a protein of SARS-CoV-2. Sci Rep. (2021) 11:13464. doi: 10.1038/s41598-021-92941-2

PubMed Abstract | Crossref Full Text | Google Scholar

345. Brasier A. The nuclear factor-kappaB-interleukin-6 signalling pathway mediating vascular inflammation. Cardiovasc Res. (2010) 86:211–8. doi: 10.1093/cvr/cvq076

PubMed Abstract | Crossref Full Text | Google Scholar

346. Hirano T, Murakami M. COVID-19: A new virus, but a familiar receptor and cytokine release syndrome. Immunity. (2020) 52:731–3. doi: 10.1016/j.immuni.2020.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

347. Murakami M, Hirano T. A four-step model for the IL-6 amplifier, a regulator of chronic inflammations in tissue-specific MHC class II-associated autoimmune diseases. Front Immunol. (2011) 2:22. doi: 10.3389/fimmu.2011.00022

PubMed Abstract | Crossref Full Text | Google Scholar

348. Suzuki H, Raska M, Yamada K, Moldoveanu Z, Julian B, Wyatt R, et al. Cytokines alter IgA1 O-glycosylation by dysregulating C1GalT1 and ST6GalNAc-II enzymes. J Biol Chem. (2014) 289:5330–9. doi: 10.1074/jbc.M113.512277

PubMed Abstract | Crossref Full Text | Google Scholar

349. Heineke M, Ballering A, Jamin A, Ben Mkaddem S, Monteiro R, Van Egmond M. New insights in the pathogenesis of immunoglobulin A vasculitis (Henoch-Schönlein purpura). Autoimmun Rev. (2017) 16:1246–53. doi: 10.1016/j.autrev.2017.10.009

PubMed Abstract | Crossref Full Text | Google Scholar

350. Rops A, Jansen E, van der Schaaf A, Pieterse E, Rother N, Hofstra J, et al. Interleukin-6 is essential for glomerular immunoglobulin A deposition and the development of renal pathology in Cd37-deficient mice. Kidney Int. (2018) 93:1356–66. doi: 10.1016/j.kint.2018.01.005

PubMed Abstract | Crossref Full Text | Google Scholar

351. Wardle E. Is IgA nephropathy induced by hyperproduction of interferon-alpha? Med Hypotheses. (2004) 62:625–8. doi: 10.1016/j.mehy.2003.11.002

PubMed Abstract | Crossref Full Text | Google Scholar

352. Sprent J, King C. COVID-19 vaccine side effects: The positives about feeling bad. Sci Immunol. (2021) 6:eabj9256. doi: 10.1126/sciimmunol.abj9256

PubMed Abstract | Crossref Full Text | Google Scholar

353. Cugno M, Meroni P, Gualtierotti R, Griffini S, Grovetti E, Torri A, et al. Complement activation in patients with COVID-19: A novel therapeutic target. J Allergy Clin Immunol. (2020) 146:215–7. doi: 10.1016/j.jaci.2020.05.006

PubMed Abstract | Crossref Full Text | Google Scholar

354. Scholz W, McClurg M, Cardenas G, Smith M, Noonan D, Hugli T, et al. C5a-mediated release of interleukin 6 by human monocytes. Clin Immunol Immunopathol. (1990) 57:297–307. doi: 10.1016/0090-122990043-p

Crossref Full Text | Google Scholar

355. Grailer J, Bosmann M, Ward P. Regulatory effects of C5a on IL-17A, IL-17F, and IL-23. Front Immunol. (2013) 3:387. doi: 10.3389/fimmu.2012.00387

PubMed Abstract | Crossref Full Text | Google Scholar

356. Anliker-Ort M, Dingemanse J, van den Anker J, Kaufmann P. Treatment of Rare inflammatory kidney diseases: Drugs targeting the terminal complement pathway. Front Immunol. (2020) 11:599417. doi: 10.3389/fimmu.2020.599417

PubMed Abstract | Crossref Full Text | Google Scholar

357. Cheng M, Zhang S, Porritt R, Arditi M, Bahar I. An insertion unique to SARS-CoV-2 exhibits superantigenic character strengthened by recent mutations. bioRxiv [Preprint]. (2020): doi: 10.1101/2020.05.21.109272

PubMed Abstract | Crossref Full Text | Google Scholar

358. Cheng M, Zhang S, Porritt R, Noval Rivas M, Paschold L, Willscher E, et al. Superantigenic character of an insert unique to SARS-CoV-2 spike supported by skewed TCR repertoire in patients with hyperinflammation. Proc Natl Acad Sci U S A. (2020) 117:25254–62. doi: 10.1073/pnas.2010722117

PubMed Abstract | Crossref Full Text | Google Scholar

359. Fraser J. Clarifying the mechanism of superantigen toxicity. PLoS Biol. (2011) 9:e1001145. doi: 10.1371/journal.pbio.1001145

PubMed Abstract | Crossref Full Text | Google Scholar

360. Koo T, Kim G, Park M. Clinicopathologic features of IgA-dominant postinfectious glomerulonephritis. Korean J Pathol. (2012) 46:105–14. doi: 10.4132/KoreanJPathol.2012.46.2.105

PubMed Abstract | Crossref Full Text | Google Scholar

361. Kuba K, Imai Y, Rao S, Gao H, Guo F, Guan B, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med. (2005) 11:875–9. doi: 10.1038/nm1267

PubMed Abstract | Crossref Full Text | Google Scholar

362. Eguchi S, Kawai T, Scalia R, Rizzo V. Understanding angiotensin II Type 1 receptor signaling in vascular pathophysiology. Hypertension. (2018) 71:804–10. doi: 10.1161/HYPERTENSIONAHA.118.10266

PubMed Abstract | Crossref Full Text | Google Scholar

363. Okamoto H, Ichikawa N. The pivotal role of the angiotensin-II-NF-κB axis in the development of COVID-19 pathophysiology. Hypertens Res. (2021) 44:126–8. doi: 10.1038/s41440-020-00560-7

PubMed Abstract | Crossref Full Text | Google Scholar

364. Masi S, Uliana M, Virdis A. Angiotensin II and vascular damage in hypertension: Role of oxidative stress and sympathetic activation. Vascul Pharmacol. (2019) 115:13–7. doi: 10.1016/j.vph.2019.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

365. South A, Brady T, Flynn J. ACE2 (Angiotensin-Converting Enzyme 2), COVID-19, and ACE Inhibitor and Ang II (Angiotensin II) receptor blocker use during the pandemic: The pediatric perspective. Hypertension. (2020) 76:16–22. doi: 10.1161/HYPERTENSIONAHA.120.15291

PubMed Abstract | Crossref Full Text | Google Scholar

366. Gaddam R, Chambers S, Bhatia M. ACE and ACE2 in inflammation: A tale of two enzymes. Inflamm Allergy Drug Targets. (2014) 13:224–34. doi: 10.2174/1871528113666140713164506

PubMed Abstract | Crossref Full Text | Google Scholar

367. Hikmet F, Méar L, Edvinsson Å, Micke P, Uhlén M, Lindskog C. The protein expression profile of ACE2 in human tissues. Mol Syst Biol. (2020) 16:e9610. doi: 10.15252/msb.20209610

PubMed Abstract | Crossref Full Text | Google Scholar

368. Bank S, De S, Bankura B, Maiti S, Das M, Khan G. ACE/ACE2 balance might be instrumental to explain the certain comorbidities leading to severe COVID-19 cases. Biosci Rep. (2021) 41:BSR20202014. doi: 10.1042/BSR20202014

PubMed Abstract | Crossref Full Text | Google Scholar

369. Ciaglia E, Vecchione C, Puca A. COVID-19 infection and circulating ACE2 levels: protective role in women and children. Front Pediatr. (2020) 8:206. doi: 10.3389/fped.2020.00206

PubMed Abstract | Crossref Full Text | Google Scholar

370. Ni J, Yang F, Huang X, Meng J, Chen J, Bader M, et al. Dual deficiency of angiotensin-converting enzyme-2 and Mas receptor enhances angiotensin II-induced hypertension and hypertensive nephropathy. J Cell Mol Med. (2020) 24:13093–103. doi: 10.1111/jcmm.15914

PubMed Abstract | Crossref Full Text | Google Scholar

371. Sodhi C, Wohlford-Lenane C, Yamaguchi Y, Prindle T, Fulton W, Wang S, et al. 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. (2018) 314:L17–31. doi: 10.1152/ajplung.00498.2016

PubMed Abstract | Crossref Full Text | Google Scholar

372. Beyerstedt S, Casaro E, Rangel É. COVID-19: Angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur J Clin Microbiol Infect Dis. (2021) 40:905–19. doi: 10.1007/s10096-020-04138-6

PubMed Abstract | Crossref Full Text | Google Scholar

373. Annweiler C, Cao Z, Wu Y, Faucon E, Mouhat S, Kovacic H, et al. Counter-regulatory ‘Renin-Angiotensin’ system-based candidate drugs to treat COVID-19 Diseases in SARS-CoV-2-infected Patients. Infect Disord Drug Targets. (2020) 20:407–8. doi: 10.2174/1871526520666200518073329

PubMed Abstract | Crossref Full Text | Google Scholar

374. Caputo I, Caroccia B, Frasson I, Poggio E, Zamberlan S, Morpurgo M, et al. Angiotensin II Promotes SARS-CoV-2 infection via upregulation of ACE2 in human bronchial cells. Int J Mol Sci. (2022) 23:5125. doi: 10.3390/ijms23095125

PubMed Abstract | Crossref Full Text | Google Scholar

375. Pinto B, Oliveira A, Singh Y, Jimenez L, Gonçalves A, Ogava R, et al. ACE2 expression is increased in the lungs of patients with comorbidities associated with severe covID-19. J Infect Dis. (2020) 222:556–63. doi: 10.1093/infdis/jiaa332

PubMed Abstract | Crossref Full Text | Google Scholar

376. Azinheira Nobrega Cruz N, Gonçalves de Oliveira L, Tedesco Silva H, Osmar Medina Pestana J, Casarini D. Angiotensin-converting enzyme 2 in the pathogenesis of renal abnormalities observed in COVID-19 patients. Front Physiol. (2021) 12:700220. doi: 10.3389/fphys.2021.700220

PubMed Abstract | Crossref Full Text | Google Scholar

377. Diebold M, Locher E, Boide P, Enzler-Tschudy A, Faivre A, Fischer I, et al. Incidence of new onset glomerulonephritis after SARS-CoV-2 mRNA vaccination is not increased. Kidney Int. (2022) 102:1409–19. doi: 10.1016/j.kint.2022.08.021

PubMed Abstract | Crossref Full Text | Google Scholar

378. Canney M, Atiquzzaman M, Cunningham A, Zheng Y, Er L, Hawken S, et al. A population-based analysis of the risk of glomerular disease relapse after COVID-19 vaccination. J Am Soc Nephrol. (2022) 33:2247–57. doi: 10.1681/ASN.2022030258

PubMed Abstract | Crossref Full Text | Google Scholar

379. Kebayoon A, Wiwanitkit V. Dengue after COVID-19 vaccination: Possible and might be missed. Clin Appl Thromb Hemost. (2021) 27:10760296211047229. doi: 10.1177/10760296211047229

PubMed Abstract | Crossref Full Text | Google Scholar

380. Mungmunpuntipantip R, Wiwanitkit V. Antineutrophil cytoplasmic antibody, glomerulonephritis and inactivated SARS-CoV-2 vaccine: Comment on the article by Garcia et al. ACR Open Rheumatol. (2022) 4:644. doi: 10.1002/acr2.11438

PubMed Abstract | Crossref Full Text | Google Scholar

381. Eslambolchi A, Aghaghazvini L, Gholamrezanezhad A, Kavosi H, Radmard A. Coronavirus disease 2019 (COVID-19) in patients with systemic autoimmune diseases or vasculitis: Radiologic presentation. J Thromb Thrombolysis. (2019) 51:339–48. doi: 10.1007/s11239-020-02289-z

PubMed Abstract | Crossref Full Text | Google Scholar

382. Becker R. COVID-19-associated vasculitis and vasculopathy. J Thromb Thrombolysis. (2020) 50:499–511. doi: 10.1007/s11239-020-02230-4

PubMed Abstract | Crossref Full Text | Google Scholar

383. Qurratulain Q, Ahmed A, Jones Q. Lesson of the month: Severe granulomatosis with polyangiitis (GPA): A diagnostic challenge during the COVID-19 pandemic. Clin Med. (2021) 21:79–80. doi: 10.7861/clinmed.2020-0793

PubMed Abstract | Crossref Full Text | Google Scholar

384. Hashizume H, Sano Y, Furukawa S, Imokawa S. Eosinophilic granulomatosis with polyangiitis mimicking coronavirus disease 2019: A case report. J Eur Acad Dermatol Venereol. (2019) 34:e557–9. doi: 10.1111/jdv.16705

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: COVID-19, glomerulonephritis, glomerulopathies, COVID-19 associated nephropathy, immunology, immune complex, cytokines, vaccines

Citation: Coyne BM, Ito D, Tariq A, Lew SQ, Kopp J, Vinales PC, Malik F, Gipson PE and Nobakht E (2025) Ascertaining the mechanistic etiology of COVID-associated glomerulonephritis: a systematic review. Front. Med. 12:1568943. doi: 10.3389/fmed.2025.1568943

Received: 31 January 2025; Accepted: 14 May 2025;
Published: 09 June 2025.

Edited by:

Vincenzo Cantaluppi, University of Eastern Piedmont, Italy

Reviewed by:

Tibor Nadasdy, The Ohio State University, United States
Kristin Meliambro, Icahn School of Medicine at Mount Sinai, United States

Copyright © 2025 Coyne, Ito, Tariq, Lew, Kopp, Vinales, Malik, Gipson and Nobakht. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Patrick E. Gipson, cGdpcHNvbkBtZmEuZ3d1LmVkdQ==

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.