Next Article in Journal
Molecular Mechanisms of MmuPV1 E6 and E7 and Implications for Human Disease
Previous Article in Journal
Proteome Analysis of Swine Macrophages after Infection with Two Genotype II African Swine Fever Isolates of Different Pathogenicity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New-Onset and Relapsed Membranous Nephropathy post SARS-CoV-2 and COVID-19 Vaccination

Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang 330006, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2022, 14(10), 2143; https://doi.org/10.3390/v14102143
Submission received: 22 August 2022 / Revised: 16 September 2022 / Accepted: 25 September 2022 / Published: 28 September 2022
(This article belongs to the Section SARS-CoV-2 and COVID-19)

Abstract

:
Since the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak and COVID-19 vaccination, new-onset and relapsed clinical cases of membranous nephropathy (MN) have been reported. However, their clinical characteristics and pathogenesis remained unclear. In this article, we collected five cases of MN associated with SARS-CoV-2 infection and 37 related to COVID-19 vaccination. Of these five cases, four (4/5, 80%) had acute kidney injury (AKI) at disease onset. Phospholipase A2 receptor (PLA2R) in kidney tissue was negative in three (3/5, 60%) patients, and no deposition of virus particles was measured among all patients. Conventional immunosuppressive drugs could induce disease remission. The underlying pathogenesis included the subepithelial deposition of viral antigens and aberrant immune response. New-onset and relapsed MN after COVID-19 vaccination generally occurred within two weeks after the second dose of vaccine. Almost 27% of patients (10/37) suffered from AKI. In total, 11 of 14 cases showed positive for PLA2R, and 20 of 26 (76.9%) presented with an elevated serum phospholipase A2 receptor antibody (PLA2R-Ab), in which 8 cases exceeded 50 RU/mL. Conventional immunosuppressive medications combined with rituximab were found more beneficial to disease remission for relapsed patients. In contrast, new-onset patients responded to conservative treatment. Overall, most patients (24/37, 64.9%) had a favorable prognosis. Cross immunity and enhanced immune response might contribute to explaining the mechanisms of MN post COVID-19 vaccination.

1. Introduction

The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has brought significant challenges to human beings. As of 11 September 2022, more than 605 million infected cases have been confirmed globally, and deaths have reached 6.4 million [1]. Current research found that SARS-CoV-2 could affect multiple human organs with a high expression of angiotensin-converting-enzyme 2 (ACE2) receptor, including respiratory tracts, heart, kidney, nervous system, etc. [2].
Admittedly, the rapid development of vaccines effectively curbed COVID-19 prevalence and transmission, including mRNA vaccines (Pfizer-BioNTech, America; Moderna, America), inactivated vaccines (Sinovac Life Sciences, China), and adenovirus vector vaccines (AstraZeneca, America; Johnson & Johnson, America), all of which relied upon and aimed to present spike protein to the immune system despite different functional patterns. Common adverse reactions after COVID-19 vaccination included fever, headache, fatigue, myalgia, etc. [3]. Nevertheless, since massive vaccination, growing numbers of clinical cases concerning glomerular diseases such as membranous nephropathy (MN) [4], IgA nephropathy [5,6], and minimal change disease [7], etc., have been widely reported.
MN was an autoimmune glomerular disease in adults, usually manifested as edema and proteinuria. Approximately one-third of patients could achieve spontaneous remission [8], and immune therapies were considered to be the preferred treatment regime, including B lymphocyte depletion, steroids, cyclophosphamide (CTX), and calcineurin inhibitors [8]. With the continuous increase of infected individuals and the universal application of COVID-19 vaccination, multiple clinical cases of new-onset and relapsed MN were reported, whereas the association between them remained mysterious. In this review, we systematically summarized the clinical features of new-onset and relapsed MN post SARS-CoV-2 infection and COVID-19 vaccination, elaborated their treatment and prognosis, and first proposed several potential mechanisms of the disease.

2. New-Onset MN post SARS-CoV-2 Infection

2.1. Clinical Features and Follow-Up

We performed a literature review via searching the electronic database, including PubMed, EMBASE, Google Scholar, and Web of Science, taking (“membranous nephropathy” OR “proteinuria” OR “nephrotic syndrome”) AND (“SARS-CoV-2” OR “COVID-19” OR “2019-ncov” OR “novel coronavirus” OR “coronavirus”) as the keywords to acquire the clinical cases of MN related to SARS-CoV-2 infection.
In total, five cases of MN associated with SARS-CoV-2 infection have been identified before 6 September 2022 in four articles [9,10,11,12] (Table 1)—all new, including one female and four males. Four elderly cases had a previous chronic medical history, especially hypertension. Three cases developed edema, and one case showed massive proteinuria. All patients underwent kidney biopsy (phospholipase A2 receptor (PLA2R)—two positive, three negative; viral particles, all negative). Only one case [11] presented with PLA2R-Ab positive expression. The auxiliary examination of four cases suggested acute kidney injury (AKI) at admission.
One elderly female case [9] saw improvement in edema without any intervention, and the other case [9] responded to tacrolimus (TAC), but his follow-up records were not available. One male case [10] died of worsened respiratory status within 16 days. Despite that, the albumin increased from 17 to 26 g/L, and serum creatinine decreased from 7.1 to 3.7 mg/dL with the treatment of lenzilumab and intravenous methylprednisolone. A young patient [11] initially received angiotensin-converting-enzyme inhibitor (ACEI), whereas there was no remission at 3 months, followed by CTX combined with prednisolone which achieved gradual remission within 2 months. One elderly male case [12] had been dependent on dialysis for 80 days.

2.2. Treatment and Prognosis

Case reports on new-onset MN post SARS-CoV-2 infection were rare. Only one case [9] achieved spontaneous remission without treatment. Steroids, CTX, and TAC were the principal immunotherapy approaches. Up to now, the published cases of MN associated with SARS-CoV-2 infection were not recommended using rituximab (RTX) (Table 1, case 1–5). Previous literature reported that RTX treatment could cause viral reactivation among patients with hepatitis B virus-associated MN [14]. Whether this phenomenon would occur in MN post SARS-CoV-2 infection still needs further clarification. In contrast, the prognosis of elderly patients with chronic kidney disease was relatively dismal compared with those without a medical history of renal involvement.
In addition, one patient with a prior diagnosis of MN infected with SARS-CoV-2 following the administration of RTX. He achieved viral elimination within 3 weeks after anti-virus medications, and no serious adverse events occurred (Table 1, case 6).

3. New-Onset and Relapsed MN post COVID-19 Vaccination

3.1. Clinical Features and Follow-Up

3.1.1. All Cases

Further, we conducted a literature search based on PubMed, EMBASE, Google Scholar, and Web of Science electronic database via the following keywords: (“membranous nephropathy” OR “proteinuria” OR “nephrotic syndrome”) AND (“SARS-CoV-2” OR “COVID-19” OR “2019-ncov” OR “novel coronavirus” OR “coronavirus”) AND (“vaccine” OR “vaccination”) to collect the clinical information of new-onset and relapsed MN post COVID-19 vaccination.
There were 37 cases reported before 6 September 2022 in 20 articles [4,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33] (Table 2), including 20 (54.1%) cases with new diagnoses and 17 (45.9%) cases with relapsed or worsening symptoms. The median age of onset was 63.5 (22–84) years, and males accounted for 67.6% (25/37). mRNA vaccines were the principal type (30/38, 78.9%), followed by adenovirus vector vaccines (5/38, 13.2%). More than half of all patients were secondary to the second dose of vaccine within two weeks. The most frequent clinical presentation was edema. There were 10 cases (10/37, 27.0%) that suffered from AKI. However, AKI occurred in 4 out of five patients with MN secondary to SARS-CoV-2 infection. Among these 14 cases with available data of PLA2R staining, 11 presented with positive expression. In total, 20 of 26 (76.9%) cases showed an elevated level of PLA2R-Ab, in which 8 cases exceeded 50 RU/mL. Most of the cases (24/37, 64.9%) were given immunosuppressive therapies, and 12 cases were treated conservatively. A total of 24 cases responded to conservative, conventional immunosuppressive medications with or without RTX. All data could be acquired in Table 3.

3.1.2. New-Onset MN

We collected 20 patients with new-onset MN post COVID-19 vaccination, and the median age of onset was 57 (22–82) years, of which 12 were males. mRNA vaccines were the leading type (15/21, 71.4%), usually occurring after the second dose of vaccine (10/20, 50.0%), with the most common onset time within two weeks (10/20, 50.0%). Edema and proteinuria were commonly observed in these cases. In total, 6 of 20 cases (30%) showed AKI. There were 7 cases associated with PLA2R, one case [4] with thrombospondin type-1 domain-containing 7A (THSD7A), and the other case [21] was diagnosed as neural epidermal growth factor-like 1 (NELL-1) related MN.
A total of 11 cases were treated with immunosuppressive drugs, 8 of which were given RTX, 4 patients achieved remission within the follow-up period, 1 case showed no response within 2 months, and 3 cases were lost to follow-up. Another 9 cases received conservative measures, 7 cases underwent remission, 1 case showed no response, and 1 case had no follow-up information. The median remission time was 41 (14–180) days. In general, the clinical treatment effect on COVID-19 vaccination-associated MN was worthy of being recognized. Table 2 and Table 3 illustrated the detailed data.

3.1.3. Relapsed or Worsening MN

Overall, 17 cases showed worsening edema and proteinuria. Of the majority of enrolled patients, 76.5% (13/17), were males, with the median age of 65 (39–84) years. A total of 15 cases were associated with mRNA vaccines, in which Pfizer-BioNTech accounted for 86.7% (13/15). Two doses of vaccines were more likely to cause disease recurrence. Among these 17 patients, 8 patients relapsed within 2 weeks. In total, 14 of 15 cases (93.3%) were represented as an elevation of PLA2R-Ab. AKI was reported in 4 cases (4/17, 23.5%).
There were 13 cases treated with immunosuppressive medications, of which three cases responded to TAC, 2 to prednisone, and 1 patient using obinutuzumab had unclear prognostic information. A total of 7 cases received RTX, only one elderly patient [29] showed no remission in 4 months. Three cases received conservative treatment, only 1 case [31] showed improvement in proteinuria. The median remission time was 58 (30–180) days. In all relapsed cases, in 3 patients [29] using immunosuppressive medications during vaccination, edema occurred (Table 2 and Table 3).

3.2. Treatment and Prognosis

Generally, clinical cases of new-onset and relapsed MN associated with COVID-19 vaccination had an excellent prognosis. Some patients, especially new-onset patients, could achieve remission via conservative management. The median remission time was 30 (14–210) days. In contrast, conventional immunosuppressive drugs combined with RTX were required for relapsed patients, and the median remission time was 60 (21–180) days. It was widely accepted that PLA2R-Ab was a crucial clinical indicator for predicting the prognosis of MN [34]. Among these 20 cases of MN secondary to COVID-19 vaccination with positive expression of PLA2R-Ab, 14 had achieved remission in the follow-up period, and the median remission time was 60 (21–180) days (Table 2).

4. Discussions

4.1. Potential Mechanisms of MN post SARS-CoV-2 Infection

4.1.1. Subepithelial Deposition of Viral Antigens

Spike protein was the pivotal structure for SARS-CoV-2 to infect host cells through specifically recognizing ACE2 [35] and with the assistance of being cleaved by transmembrane protease serine 2 (TMPRSS2) [36,37]. Notably, both ACE2 and TMPRSS2 were also expressed on podocytes [38]. According to reports in the literature, SARS-CoV-2 viral particles were detected in podocytes of postmortem kidney samples in 26 patients with COVID-19 [39], indicating potential evidence that SARS-CoV-2 could directly invade into podocytes. Nevertheless, the process of viral infection on podocytes might contribute to the subepithelial deposition of viral antigens [40], thus forming in situ antigens to stimulate the production of corresponding antibodies, leading to the deposition of viral immune complexes in glomeruli (Figure 1).

4.1.2. Massive Release of Cytokines

In COVID-19, viral infection in pulmonary epithelial cells triggered the recruitment of immune effector cells and released massive proinflammatory cytokines and chemokines [41,42], which subsequently advanced T lymphocyte differentiation. T helper (Th) 17 cells generated interleukin (IL)-17A, IL-17F, IL-22, and granulocyte-macrophage colony-stimulating factor, inducing the aggregation of inflammatory cells, such as neutrophils [43]. IL-4 produced by Th2 cells and IL-21 produced by follicular T helper (Tfh) cells contributed to B lymphocyte survival and proliferation as well as generated higher affinity to the IgG4 antibody [44]. Moreover, IL-4, IL-13, and IL-10 could promote the conversion of antibody category to IgG4 [44]. In addition, decreased levels of Th1 cells and regulatory T (Treg) cells secondary to SARS-CoV-2 infection destroyed immune tolerance [45].
In addition to acting as receptors, ACE2 also presented critical functions as a counter-regulatory enzyme to convert angiotensin II (Ang II) into Ang-(1–7), the latter of which performed attenuating inflammation effects [46]. Virus-occupied ACE2 might weaken their intrinsic function, which could enhance inflammatory response, neutrophils accumulation, and vascular permeability, and ultimately result in influenza-like symptoms, even severe acute respiratory distress syndrome among SARS-CoV-2 infected individuals [47], whereas in the kidneys, elevated immune response made it easier to develop glomerular diseases, such as MN (Figure 1).

4.1.3. Speculation about PLA2R Antigen

Accumulating evidence has demonstrated that PLA2R, a pathogenic antigen of MN, was expressed not only in airway epithelial cells [48], neutrophils [49], and pulmonary macrophages [50], but also in podocytes. Once activated by foreign antigens such as SARS-CoV-2, these cells could secret extracellular vesicles containing PLA2R or cause the spatial release of PLA2R by generating extracellular traps, subsequently stimulating B lymphocytes to produce PLA2R-Ab [51]. In addition, the oxidation environment induced by inflammatory cytokines could bring about long-term expression of PLA2R pathogenic epitopes and enhance the capacity of binding to circulating antibodies [51] (Figure 1).

4.1.4. Activation of the Complement System

The previous literature reported that the concentration of anti-SARS-CoV-2 immunoglobulin lacking glycan fucosylation was elevated in COVID-19 patients [52], which could help mannose-binding lectin to combine with aberrant glycans, thereby activating the complement system. The formed C5b-9 membrane attack complex on podocyte membranes participated in mediating the proteolysis of podocyte synaptophysin and NEPH1, resulting in the destruction of podocyte cytoskeleton [53,54] and eventually proteinuria (Figure 1).

4.1.5. Elevated Expression of Human Leukocyte Antigen

Human leukocyte antigen (HLA) was expressed on the surface of immune cells and acted as presenters of epitopes to CD4+ T cells [55], indicating potent immunoregulatory properties. The present study demonstrated that activation of HLA-DR in circulating monocytes increased instantaneously in patients with SARS-CoV-2 infection [56], which could promote antigenic epitopes presentation to T lymphocytes and destroy immune tolerance (Figure 1).

4.2. Potential Mechanisms of MN post COVID-19 Vaccination

4.2.1. Cross Immune Response

Some scholars have proven that amino acid sequence similarity between hepatitis B virus surface antigen and multiple sclerosis (MS) autoantigens might be part of the account of MS secondary to hepatitis B vaccination [57]. Recently, Vojdani et al. conducted an investigation aimed at studying the relationship between autoimmune target proteins and SARS-CoV-2 spike protein antibodies, the results of which proved that there were multiple tissue antigens that showed powerful reactions with the SARS-CoV-2 antibodies, such as transglutaminase 3, anti-extractable nuclear antigen, thyroid peroxidase, etc. [58], which indicated the fundamental role of cross-immune response in autoimmune diseases. Therefore, we proposed an underlying mechanism that podocyte surface-specific antigens might share similar amino acid sequences with spike protein or other components of SARS-CoV-2, which would be further supported along with the discovery of more pathogenic antigens on the surface of podocytes (Figure 2).

4.2.2. Subepithelial Deposition of Circulating Immune Complexes

Proverbially, intramuscular vaccine components served as foreign substances to evoke the host’s immune response. As antigens, the vaccines promoted subepithelial deposition of circulating immune complexes in renal tissue via combining with native antibodies in vivo, which was plausible for explaining the MN secondary to influenza vaccines [59] (Figure 2).

4.2.3. Enhanced Immune Response

Anti-SARS-CoV-2 neutralizing antibodies were of particular significance in evaluating protective immunity. For 250 patients with past-COVID-19, all moderate-severe patients and more than 80% of mild patients had positive antibodies [60]. Undeniably, vaccination was indeed an important initiative to enhance immune response. Compared with other types of COVID-19 vaccines, mRNA vaccines have been revealed to induce a more potent immune response. The rate of seroconversion was observed to increase five-fold from the baseline after the first dose of mRNA vaccines at two weeks [60]. Two doses of vaccines effectively induced antibody titers to exceed 300 U/mL and without evident decrease at 2 months [60]. Vaccines strengthened virus-specific responses and effectively activated T and B lymphocytes, accompanied by elevated generation of T cell inflammatory cytokines (e.g., interferon γ, tumor necrosis factor α, and IL-2, etc.) and higher levels of antibody titers, especially two doses of vaccines, including elderly individuals [61,62]. Due to the dose-dependent characteristics of COVID-19 vaccines, most adverse immune events typically occurred post the second dose [63] (Figure 2), which was consistent with our result. According to the clinical records we collected, only four new-onset patients had available data on antibody titers (case 2, 147.0 U/mL; case5, 32.8 U/mL; case19, 2334 U/mL; case 20, 1500 U/mL, Table 2). Consequently, further studies regarding the detection of antibody titers were required, which might contribute to clarifying the correlation between antibody level and disease onset.

4.2.4. Adjuvants

The application of adjuvants achieved the possibility that small doses of vaccines could stimulate individuals to generate sufficient antibodies. Adjuvants, as antigens, could provide pathogen-associated molecular patterns and be recognized by toll-like receptors on the surface of antigen-presenting cells, activating downstream inflammatory signaling pathways and inducing an enhanced immune response [64]. Inactivated vaccines had decreased immunogenicity and usually required adjuvants, especially in elderly individuals considering immune senescence [65] (Figure 2).

4.3. Limitations

This article had several limitations. Most clinical cases were reported from single-case studies, and further research was needed to verify the causal relationship between MN and SARS-CoV-2 infection and COVID-19 vaccination. Moreover, there was the possibility that multiple cases were not reported, which might perplex us concerning the clinical characteristics and treatment response of MN associated with SARS-CoV-2 infection and COVID-19 vaccination. Furthermore, the pathogenesis involved in this review was based on our hypothesis, which demanded additional verification in the future. In addition, insufficient clinical information might lead to errors in data analysis.

5. Conclusions

In the era of the COVID-19 pandemic and the widespread requirement for vaccines, MN was an unavoidable but uncommon disease. Edema and proteinuria were the leading clinical manifestations. Overall, most cases had a good prognosis. Conservative and conventional immunosuppressive therapies with or without RTX promoted disease remission. Of note, it was not emphasized enough on the detection of antibody titers in currently published cases of MN post COVID-19 vaccination, which would be meaningful for assessing the potential association between antibody levels and disease onset. In addition, whether routine urine testing after vaccination contributed to the timely detection of the disease deserved more attention. In conclusion, further exploration was urgently needed to advance our knowledge of the incidence and recurrence rate, pathogenesis, treatment, and prognosis of MN post SARS-CoV-2 infection and COVID-19 vaccination.

Author Contributions

Q.M. conducted data collection and wrote the manuscript. X.L. conducted data analysis. G.X. was responsible for the idea, funds, and paper revision. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Natural Science Foundation of China (No. 81970583 & 82060138), the Nature Science Foundation of Jiangxi Province (No. 20202BABL206025), and the Kidney Disease Engineering Technology Research Centre Foundation of Jiangxi Province (No. 20164BCD40095).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACE2: angiotensin-converting enzyme 2; ACEI, angiotensin-converting enzyme inhibitors; Ang II, angiotensin II; AKI, acute kidney disease; COVID-19, coronavirus disease 2019; CTX, cyclophosphamide; IL, interleukin; MN, membranous nephropathy; MS, multiple sclerosis; NELL-1, neural epidermal growth factor-like 1; PLA2R, phospholipase A2 receptor; PLA2R-Ab, phospholipase A2 receptor antibody; RTX, rituximab; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TAC, tacrolimus; Tfh, follicular T helper; Th, T helper; THSD7A, thrombospondin type-1 domain-containing 7A; TMPRSS2, transmembrane protease serine 2; Treg, regulatory T.

References

  1. World Health Organization. COVID-19 Weekly Epidemiological Update. Available online: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/situation-reports (accessed on 16 September 2022).
  2. Tuttolomondo, D.; Frizzelli, A.; Aiello, M.; Bertorelli, G.; Majori, M.; Chetta, A. Beyond the lung involvement in COVID-19 patients. Minerva Med. 2022, 113, 558–568. [Google Scholar] [CrossRef] [PubMed]
  3. Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416. [Google Scholar] [CrossRef] [PubMed]
  4. Da, Y.; Goh, G.H.; Khatri, P. A case of membranous nephropathy following Pfizer-BioNTech mRNA vaccination against COVID-19. Kidney Int. 2021, 100, 938–939. [Google Scholar] [CrossRef] [PubMed]
  5. Rahim, S.E.G.; Lin, J.T.; Wang, J.C. A case of gross hematuria and IgA nephropathy flare-up following SARS-CoV-2 vaccination. Kidney Int. 2021, 100, 238. [Google Scholar] [CrossRef] [PubMed]
  6. Ma, Y.; Xu, G. New-Onset IgA nephropathy Following COVID-19 Vaccination. QJM Int. J. Med. 2022, hcac185. [Google Scholar] [CrossRef]
  7. Leclerc, S.; Royal, V.; Lamarche, C.; Laurin, L.P. Minimal Change Disease With Severe Acute Kidney Injury Following the Oxford-AstraZeneca COVID-19 Vaccine: A Case Report. Am. J. Kidney Dis. 2021, 78, 607–610. [Google Scholar] [CrossRef]
  8. Couser, W.G. Primary Membranous Nephropathy. Clin. J. Am. Soc. Nephrol. 2017, 12, 983–997. [Google Scholar] [CrossRef]
  9. Kudose, S.; Batal, I.; Santoriello, D.; Xu, K.; Barasch, J.; Peleg, Y.; Canetta, P.; Ratner, L.E.; Marasa, M.; Gharavi, A.G.; et al. Kidney Biopsy Findings in Patients with COVID-19. J. Am. Soc. Nephrol. 2020, 31, 1959–1968. [Google Scholar] [CrossRef]
  10. Miao, J.; Fidler, M.E.; Nasr, S.H.; Larsen, C.P.; Zoghby, Z.M. Membranous nephropathy in a patient with coronavirus disease 2019 (COVID-19): A case report. Clin. Nephrol. Case Stud. 2021, 9, 11–18. [Google Scholar] [CrossRef]
  11. Guo, W.; Tan, P.H.; Baikunje, S. Membranous nephropathy in a patient with COVID-19 infection. J. Nephrol. 2022, 35, 351–355. [Google Scholar] [CrossRef]
  12. Nasr, S.H.; Alexander, M.P.; Cornell, L.D.; Herrera, L.H.; Fidler, M.E.; Said, S.M.; Zhang, P.; Larsen, C.P.; Sethi, S. Kidney Biopsy Findings in Patients With COVID-19, Kidney Injury, and Proteinuria. Am. J. Kidney Dis. 2021, 77, 465–468. [Google Scholar] [CrossRef] [PubMed]
  13. Elcioglu, O.C.; Artan, A.S.; Mirioglu, S.; Gursu, M.; Durdu, B.; Koc, M.M.; Okyaltirik, F.; Gultekin, M.A.; Kazancioglu, R. COVID-19 infection in a membranous nephropathy patient treated with rituximab. CEN Case Rep. 2021, 10, 83–87. [Google Scholar] [CrossRef]
  14. Berchtold, L.; Zanetta, G.; Dahan, K.; Mihout, F.; Peltier, J.; Guerrot, D.; Brochériou, I.; Ronco, P.; Debiec, H. Efficacy and Safety of Rituximab in Hepatitis B Virus-Associated PLA2R-Positive Membranous Nephropathy. Kidney Int. Rep. 2018, 3, 486–491. [Google Scholar] [CrossRef] [PubMed]
  15. Gueguen, L.; Loheac, C.; Saidani, N.; Khatchatourian, L. Membranous nephropathy following anti-COVID-19 mRNA vaccination. Kidney Int. 2021, 100, 1140–1141. [Google Scholar] [CrossRef] [PubMed]
  16. Rashid, W.; Mousa, H.; Khan, J.; Ijaz, F.; Ezell, G.D. A Case of Membranous Nephropathy Hypothesized to be Associated With COVID-19 Vaccine. Cureus 2022, 14, e24245. [Google Scholar] [CrossRef]
  17. Psyllaki, A.; Stavrakaki, I.; Androvitsanea, A.; Gakiopoulou, H.; Petrakis, I.; Stylianou, K. Two cases of glomerular involvement after vaccination against COVID-19: Epiphenomenon or causality? Clin. Kidney J. 2022, 15, 574–575. [Google Scholar] [CrossRef]
  18. Zavala-Miranda, M.F.; González-Ibarra, S.G.; Pérez-Arias, A.A.; Uribe-Uribe, N.O.; Mejia-Vilet, J.M. New-onset systemic lupus erythematosus beginning as class v lupus nephritis after COVID-19 vaccination. Kidney Int. 2021, 100, 1340–1341. [Google Scholar] [CrossRef]
  19. Caza, T.N.; Cassol, C.A.; Messias, N.; Hannoudi, A.; Haun, R.S.; Walker, P.D.; May, R.M.; Seipp, R.M.; Betchick, E.J.; Amin, H.; et al. Glomerular Disease in Temporal Association with SARS-CoV-2 Vaccination: A Series of 29 Cases. Kidney360 2021, 2, 1770–1780. [Google Scholar] [CrossRef]
  20. Zhao, Y.; Zhang, L.; Wang, G.; Guan, J.; Pai, P. The Development of De novo Acute Tubulointerstitial Nephritis and Membranous Nephropathy Following Inactivated COVID-19 Vaccine: Causal or Casual? Clin. Case Rep. Int. 2022, 6, 1344. [Google Scholar]
  21. Klomjit, N.; Alexander, M.P.; Fervenza, F.C.; Zoghby, Z.; Garg, A.; Hogan, M.C.; Nasr, S.H.; Minshar, M.A.; Zand, L. COVID-19 Vaccination and Glomerulonephritis. Kidney Int. Rep. 2021, 6, 2969–2978. [Google Scholar] [CrossRef]
  22. Fenoglio, R.; Lalloni, S.; Marchisio, M.; Oddone, V.; De Simone, E.; Del Vecchio, G.; Sciascia, S.; Roccatello, D. New Onset Biopsy-Proven Nephropathies after COVID Vaccination. Am. J. Nephrol. 2022, 53, 325–330. [Google Scholar] [CrossRef] [PubMed]
  23. Abid, S.; Najjar, Y.; Beers, K.; Mehta, S. A case of MPO-ANCA vasculitis with membranous nephropathy after COVID vaccine. Am. J. Kidney Dis. 2022, 79, S12. [Google Scholar]
  24. Marega, A.; Pizzolitto, S.; Pian, M.; Biasi, C.; Giacomini, A.; Bertoni, M.; Romano, G.; Boscutti, G. MO230: De Novo Double Glomerulopathy (Membranous Nephropathy, Mn and Collapsing Focal Segmental Glomerulosclerosis, CFSGS) Associated to Positive Myeloperoxidase-O (MPO) Antibody Following Pfizer-Biontech MRNA Vaccination COVID-19. Nephrol. Dial. Transplant. 2022, 37, i156–i1579. [Google Scholar] [CrossRef]
  25. Fornara, L.; Musetti, C.; Guglielmetti, G.; Cantaluppi, V. MO208: De Novo Glomerulonephritides Following BNT162B2 COVID-19 Vaccine: A Case Series. Nephrol. Dial. Transpl. 2022, 37, i142. [Google Scholar] [CrossRef]
  26. Jhaveri, K.; Bijol, V.; Wanchoo, R.; Keller, K.; Alexander, M.; Weins, A. COVID-19 mRNA vaccine-associated autoimmunity presenting as minimal change disease and membranous nephropathy. J. Am. Soc. Nephrol. 2021, 32, 94. [Google Scholar]
  27. Paxton, L.; McMahon, L.; Wong, L. De novo PLA2R positive membranous nephropathy following BNT162b2 mRNA COVID-19 vaccine. Intern. Med. J. 2022. [Google Scholar] [CrossRef]
  28. Saigal, M.; Taduri, G.; Gudditi, S.; Herur, S.; Alaparthi, P.; Kinjarapu, S. POS-872 Post covid vaccination- new onset glomerulonephritis- a mere co incidence or a impending reality?? Kidney Int. Rep. 2022, 7, S377. [Google Scholar] [CrossRef]
  29. Visch, R.; Wetzels, J.; Vink, C.; van de Logt, A.-E. COVID-19 vaccination in patients with membranous nephropathy. Kidney Int. Rep. 2022, 7, 1922–1923. [Google Scholar] [CrossRef]
  30. Martinez-Pitre, P.J.; Fogo, A.B.; Alqudsi, M. Relapse of Membranous Nephropathy and Renal Sarcoidosis Following Exposure to SARS-CoV-2 Vaccine (Johnson & Johnson). Am. J. Kidney Dis. 2022, 79, S102–S103. [Google Scholar]
  31. Tsuew, Y.K.; Thong, K.M.; Lee, Y.Y.; Ramanaidu, S.; Loh, C.L. POS-903 Adverse Events Following Immunisation (AEFI) of COVID-19 Vaccines in Kidney Patients - A Case Series. Kidney Int. Rep. 2022, 7, S392. [Google Scholar] [CrossRef]
  32. Aydın, M.F.; Yıldız, A.; Oruç, A.; Sezen, M.; Dilek, K.; Güllülü, M.; Yavuz, M.; Ersoy, A. Relapse of primary membranous nephropathy after inactivated SARS-CoV-2 virus vaccination. Kidney Int. 2021, 100, 464–465. [Google Scholar] [CrossRef] [PubMed]
  33. Liang, K.V.; Kurtz, E.C.; Pittappilly, M.; Ahmad, S.; Minervini, M.I. Primary membranous nephropathy flare after COVID-19 vaccination. J. Am. Soc. Nephrol. 2021, 32, 485. [Google Scholar]
  34. Kim, Y.G.; Choi, Y.-W.; Kim, S.-Y.; Moon, J.Y.; Ihm, C.-G.; Lee, T.W.; Jeong, K.H.; Yang, S.H.; Kim, Y.S.; Oh, Y.J.; et al. Anti-Phospholipase A2 Receptor Antibody as Prognostic Indicator in Idiopathic Membranous Nephropathy. Am. J. Nephrol. 2015, 42, 250–257. [Google Scholar] [CrossRef]
  35. Mousavizadeh, L.; Ghasemi, S. Genotype and phenotype of COVID-19: Their roles in pathogenesis. J. Microbiol. Immunol. Infect. 2021, 54, 159–163. [Google Scholar] [CrossRef] [PubMed]
  36. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; 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.e278. [Google Scholar] [CrossRef] [PubMed]
  37. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
  38. Lely, A.T.; Hamming, I.; van Goor, H.; Navis, G.J. Renal ACE2 expression in human kidney disease. J. Pathol. 2004, 204, 587–593. [Google Scholar] [CrossRef]
  39. Su, H.; Yang, M.; Wan, C.; Yi, L.-X.; Tang, F.; Zhu, H.-Y.; Yi, F.; Yang, H.-C.; Fogo, A.B.; Nie, X.; et al. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020, 98, 219–227. [Google Scholar] [CrossRef]
  40. May, R.M.; Cassol, C.; Hannoudi, A.; Larsen, C.P.; Lerma, E.V.; Haun, R.S.; Braga, J.R.; Hassen, S.I.; Wilson, J.; VanBeek, C.; et al. A multi-center retrospective cohort study defines the spectrum of kidney pathology in Coronavirus 2019 Disease (COVID-19). Kidney Int. 2021, 100, 1303–1315. [Google Scholar] [CrossRef]
  41. Ye, Q.; Wang, B.; Mao, J. The pathogenesis and treatment of the `Cytokine Storm’ in COVID-19. J. Infect. 2020, 80, 607–613. [Google Scholar] [CrossRef]
  42. Pasrija, R.; Naime, M. The deregulated immune reaction and cytokines release storm (CRS) in COVID-19 disease. Int. Immunopharmacol. 2021, 90, 107225. [Google Scholar] [CrossRef] [PubMed]
  43. Lee, G.R. The Balance of Th17 versus Treg Cells in Autoimmunity. Int. J. Mol. Sci. 2018, 19, 730. [Google Scholar] [CrossRef] [PubMed]
  44. Zhao, Q.; Dai, H.; Liu, X.; Jiang, H.; Liu, W.; Feng, Z.; Zhang, N.; Gao, Y.; Dong, Z.; Zhou, X.; et al. Helper T Cells in Idiopathic Membranous Nephropathy. Front. Immunol. 2021, 12, 665629. [Google Scholar] [CrossRef]
  45. Meckiff, B.J.; Ramírez-Suástegui, C.; Fajardo, V.; Chee, S.J.; Kusnadi, A.; Simon, H.; Eschweiler, S.; Grifoni, A.; Pelosi, E.; Weiskopf, D.; et al. Imbalance of Regulatory and Cytotoxic SARS-CoV-2-Reactive CD4+ T Cells in COVID-19. Cell 2020, 183, 1340–1353. [Google Scholar] [CrossRef]
  46. South, A.M.; Tomlinson, L.; Edmonston, D.; Hiremath, S.; Sparks, M.A. Controversies of renin-angiotensin system inhibition during the COVID-19 pandemic. Nat. Reviews. Nephrol. 2020, 16, 305–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Asrani, P.; Hassan, M.I. SARS-Cov-2 mediated lung inflammatory responses in host: Targeting the cytokine storm for therapeutic interventions. Mol. Cell. Biochem. 2021, 476, 675–687. [Google Scholar] [CrossRef] [PubMed]
  48. Nolin, J.D.; Ogden, H.L.; Lai, Y.; Altemeier, W.A.; Frevert, C.W.; Bollinger, J.G.; Naika, G.S.; Kicic, A.; Stick, S.M.; Lambeau, G.; et al. Identification of Epithelial Phospholipase A2 Receptor 1 as a Potential Target in Asthma. Am. J. Respir. Cell Mol. Biol. 2016, 55, 825–836. [Google Scholar] [CrossRef] [PubMed]
  49. Silliman, C.C.; Moore, E.E.; Zallen, G.; Gonzalez, R.; Johnson, J.L.; Elzi, D.J.; Meng, X.; Hanasaki, K.; Ishizaki, J.; Arita, H.; et al. Presence of the M-type sPLA2 receptor on neutrophils and its role in elastase release and adhesion. Am. J. Physiology. Cell Physiol. 2002, 283, C1102–C1113. [Google Scholar] [CrossRef]
  50. Granata, F.; Petraroli, A.; Boilard, E.; Bezzine, S.; Bollinger, J.; Del Vecchio, L.; Gelb, M.H.; Lambeau, G.; Marone, G.; Triggiani, M. Activation of cytokine production by secreted phospholipase A2 in human lung macrophages expressing the M-type receptor. J. Immunol. 2005, 174, 464–474. [Google Scholar] [CrossRef]
  51. Liu, W.; Gao, C.; Dai, H.; Zheng, Y.; Dong, Z.; Gao, Y.; Liu, F.; Zhang, Z.; Liu, Z.; Liu, W.; et al. Immunological Pathogenesis of Membranous Nephropathy: Focus on PLA2R1 and Its Role. Front. Immunol. 2019, 10, 1809. [Google Scholar] [CrossRef]
  52. Chakraborty, S.; Gonzalez, J.; Edwards, K.; Mallajosyula, V.; Buzzanco, A.S.; Sherwood, R.; Buffone, C.; Kathale, N.; Providenza, S.; Xie, M.M.; et al. Proinflammatory IgG Fc structures in patients with severe COVID-19. Nat. Immunol. 2021, 22, 67–73. [Google Scholar] [CrossRef] [PubMed]
  53. Haddad, G.; Lorenzen, J.M.; Ma, H.; de Haan, N.; Seeger, H.; Zaghrini, C.; Brandt, S.; Kölling, M.; Wegmann, U.; Kiss, B.; et al. Altered glycosylation of IgG4 promotes lectin complement pathway activation in anti-PLA2R1-associated membranous nephropathy. J. Clin. Invest. 2021, 131, e140453. [Google Scholar] [CrossRef] [PubMed]
  54. Nangaku, M.; Shankland, S.J.; Couser, W.G. Cellular response to injury in membranous nephropathy. J. Am. Soc. Nephrol. 2005, 16, 1195–1204. [Google Scholar] [CrossRef]
  55. Mladkova, N.; Kiryluk, K. Genetic Complexities of the HLA Region and Idiopathic Membranous Nephropathy. J. Am. Soc. Nephrol. 2017, 28, 1331–1334. [Google Scholar] [CrossRef] [PubMed]
  56. Schulte-Schrepping, J.; Reusch, N.; Paclik, D.; Baßler, K.; Schlickeiser, S.; Zhang, B.; Krämer, B.; Krammer, T.; Brumhard, S.; Bonaguro, L.; et al. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell 2020, 182, 1419–1440. [Google Scholar] [CrossRef]
  57. Bogdanos, D.-P.; Smith, H.; Ma, Y.; Baum, H.; Mieli-Vergani, G.; Vergani, D. A study of molecular mimicry and immunological cross-reactivity between hepatitis B surface antigen and myelin mimics. Clin. Dev. Immunol. 2005, 12, 217–224. [Google Scholar] [CrossRef]
  58. 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. [Google Scholar] [CrossRef]
  59. Patel, C.; Shah, H.H. Membranous nephropathy and severe acute kidney injury following influenza vaccination. Saudi J. Kidney Dis. Transplant. 2015, 26, 1289–1293. [Google Scholar] [CrossRef]
  60. Favresse, J.; Gillot, C.; Di Chiaro, L.; Eucher, C.; Elsen, M.; Van Eeckhoudt, S.; David, C.; Morimont, L.; Dogné, J.-M.; Douxfils, J. Neutralizing antibodies in COVID-19 patients and vaccine recipients after two doses of BNT162b2. Viruses 2021, 13, 1364. [Google Scholar] [CrossRef]
  61. Verbeke, R.; Lentacker, I.; De Smedt, S.C.; Dewitte, H. The dawn of mRNA vaccines: The COVID-19 case. J. Control. Release 2021, 333, 511–520. [Google Scholar] [CrossRef]
  62. Jadali, Z. Renal Complications Following COVID-19 Vaccination. Urol. J. 2021, 7086. [Google Scholar] [CrossRef]
  63. Sahin, U.; Muik, A.; Derhovanessian, E.; Vogler, I.; Kranz, L.M.; Vormehr, M.; Baum, A.; Pascal, K.; Quandt, J.; Maurus, D.; et al. COVID-19 vaccine BNT162b1 elicits human antibody and T(H)1 T cell responses. Nature 2020, 586, 594–599. [Google Scholar] [CrossRef] [PubMed]
  64. Scheel, B.; Braedel, S.; Probst, J.; Carralot, J.-P.; Wagner, H.; Schild, H.; Jung, G.; Rammensee, H.-G.; Pascolo, S. Immunostimulating capacities of stabilized RNA molecules. Eur. J. Immunol. 2004, 34, 537–547. [Google Scholar] [CrossRef] [PubMed]
  65. Ciabattini, A.; Nardini, C.; Santoro, F.; Garagnani, P.; Franceschi, C.; Medaglini, D. Vaccination in the elderly: The challenge of immune changes with aging. Semin. Immunol. 2018, 40, 83–94. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Potential mechanisms of MN post SARS-CoV-2 infection. (1) SARS-CoV-2 directly infected podocytes through binding to ACE2 on the surface of podocytes, which contributed to viral subepithelial deposition and resulted in functional ACE2 decreased, forming in situ immune complexes and promoting inflammatory response. (2) Viral infection initiated the innate immune response via recruiting inflammatory cells. Certain inflammatory cells probably functioned to express PLA2R. Once activated, these cells could release PLA2R antigen and stimulate B lymphocytes to produce PLA2R-Ab. (3) The activation of the innate and adaptive immune system promoted the release of inflammatory cytokines, which induced T and B lymphocyte differentiation, and the generation of antibodies. (4) SARS-CoV-2 activated the complement system, causing podocyte injury and further release of inflammatory cytokines. (5) Elevated levels of HLA-DR in monocytes promoted antigen presentation to the innate and adaptive immune system, leading to enhanced immune response.
Figure 1. Potential mechanisms of MN post SARS-CoV-2 infection. (1) SARS-CoV-2 directly infected podocytes through binding to ACE2 on the surface of podocytes, which contributed to viral subepithelial deposition and resulted in functional ACE2 decreased, forming in situ immune complexes and promoting inflammatory response. (2) Viral infection initiated the innate immune response via recruiting inflammatory cells. Certain inflammatory cells probably functioned to express PLA2R. Once activated, these cells could release PLA2R antigen and stimulate B lymphocytes to produce PLA2R-Ab. (3) The activation of the innate and adaptive immune system promoted the release of inflammatory cytokines, which induced T and B lymphocyte differentiation, and the generation of antibodies. (4) SARS-CoV-2 activated the complement system, causing podocyte injury and further release of inflammatory cytokines. (5) Elevated levels of HLA-DR in monocytes promoted antigen presentation to the innate and adaptive immune system, leading to enhanced immune response.
Viruses 14 02143 g001
Figure 2. Potential mechanisms of MN post COVID-19 vaccination. (1) Spike or other structural proteins shared many amino acid sequences with human tissue proteins. Non-specific cross-immune response increased the risk of antibody binding to podocyte pathogenic antigens. (2) Vaccines activated immune effector cells, causing T lymphocyte differentiation and releasing massive inflammatory cytokines, which subsequently induced an enhanced immune response. (3) Vaccine acted as antigens, binding to native antibodies in vivo to form circulating immune complexes and deposited in the glomeruli. (4) Adjuvants provided pathogen-associated molecular patterns (PAMPs) and were recognized by toll-like receptors (TLRs) on the surface of antigen-presenting cells (APC) to elevate inflammatory response.
Figure 2. Potential mechanisms of MN post COVID-19 vaccination. (1) Spike or other structural proteins shared many amino acid sequences with human tissue proteins. Non-specific cross-immune response increased the risk of antibody binding to podocyte pathogenic antigens. (2) Vaccines activated immune effector cells, causing T lymphocyte differentiation and releasing massive inflammatory cytokines, which subsequently induced an enhanced immune response. (3) Vaccine acted as antigens, binding to native antibodies in vivo to form circulating immune complexes and deposited in the glomeruli. (4) Adjuvants provided pathogen-associated molecular patterns (PAMPs) and were recognized by toll-like receptors (TLRs) on the surface of antigen-presenting cells (APC) to elevate inflammatory response.
Viruses 14 02143 g002
Table 1. Summary of published cases information of MN post SARS-CoV-2 infection.
Table 1. Summary of published cases information of MN post SARS-CoV-2 infection.
CaseAge/SexCountryMedical HistoryTime after DiagnosisSymptomsKidney BiopsyLaboratory Characteristics
before Treatment
TreatmentFUOutcomeOthersRef.
PLA2RVPUPROALBPLA2R-Ab
New-Onset MN
170/FAmericaHT, CAD, PVD, cervical carcinoma, GERD, HLD, obesityUNCough, fever, dyspnea, edema, AKINegNegPCR:6.8 g/g30 g/LUNNo35 dR
PCR:5–6g/g
-[9]
272/MAmericaHT, DM, HLD, gout, spinal stenosis, atrial fibrillationUNCough, pleural effusion, edemaPosNegPCR:8.8 g/g17 g/LUNTAC18 dUNRepeated COVID-19
positive
[9]
381/MSpanishProstate Ca, prediabetes, HLD, HT, CKD stage 3, AVS, CR6 dFatigue, dyspnea,
myalgia, sore throat, dry cough, poor appetite, loss of smell and taste, nausea, diarrhea, AKI, urinary
incontinence, proteinuria
NegNeg4.6 g/d17 g/LNegLenzilumab,
antibiotics, mPSL, heparin, dialysis
16 dDeath
ALB:26 g/L when 7d on admission
Scr:3.7 mg/dL hematuria[10]
429/MSouth AsianUN4 WFever, myalgia, edema, AKIPos
(weak)
Neg8.7 g/d
PCR:7.5 g/g
22 g/LPosACEI3 MoNR
PCR:11.9 g/g
ALB:31 g/L
-[11]
PCR:9.2 g/g23 g/LUNCTX, PSL2 MoR
PCR:4.9 g/g ALB:29 g/L
571/MAmericaHT, obesity, CKDUNDyspnea, fever, cough, nausea, proteinuria, AKINegNeg14.0 g/d27 g/LUNDialysis80 dNR
Dialysis
dependent
Scr:5.7 mg/dL[12]
RTX-treated MN infected SARS-CoV-2
648/MTurkeyDM, MNNACough, fever, headacheUNNAUN19 g/LPosOseltamivir,
moxifloxacin, HCQ,
azithromycin,
lopinavir/ritonavir
3 WRInfection within 1 Mo post RTX treatment[13]
Abbreviations: ACEI, angiotensin-converting-enzyme inhibitor; AKI, acute kidney injury; ALB, albumin; AVS, aortic valve stenosis; Ca, cancer; CAD, coronary artery disease; CKD, chronic kidney disease; CR, cervical radiculopathy; CTX, cyclophosphamide; d, day; DM, diabetes mellitus; F, female; FU, follow-up; GERD, gastroesophageal reflux disease; HCQ, hydroxychloroquine; HLD, hyperlipidemia; HT, hypertension; M, male; MN, membranous nephropathy; Mo, month; mPSL, methylprednisolone; NA, not applicable; Neg, negative; NR, no response; PCR, protein–creatinine ratio; PLA2R, phospholipase A2 receptor; PLA2R-Ab, phospholipase A2 receptor antibody; Pos, positive; PSL, prednisolone; PVD, peripheral vascular disease; R, response; RTX, rituximab; Scr, serum creatinine; TAC, tacrolimus; UN, unknown; UPRO, urine protein; VP, viral particles; W, week.
Table 2. Summary of published cases information of MN post COVID-19 vaccination.
Table 2. Summary of published cases information of MN post COVID-19 vaccination.
CaseAge/SexRace/
Country
Medical HistoryVaccineWhich DoseTimeSymptomsPLA2RBaseline ScrLaboratory Characteristics before TreatmentTreatmentFUOutcomeOthersRef.
UPROALBScrPLA2R-Ab
New-Onset MN
176/MFranceHT, UV-treated cutaneous mycosis
fungoid
mRNA
(Pfizer-BioNTech)
14 dEdemaNANormalPCR:
6.5 g/g
16 g/L0.9 mg/dL1:800RASB3 WR
PCR: 3.0 g/g
ALB: 26 g/L
Hematuria[15]
mRNA
(Moderna)
12 dEdema, AKINAPCR:
3.8 g/g
22 g/L1.2 mg/dLUNRTX2 MoR
ALB: elevated
270/MSingaporeNomRNA
(Pfizer-BioNTech)
11 W,
1 d after 2nd dose
Edema, AKINegNormal4.4 g/d17 g/L1.3 mg/dLNegIrbesartan,
furosemide,
warfarin
2 MoNRTHSD7A-Ab: Pos COVID-19 antibody: 147.0 U/mL[4]
356/MAmericaHTmRNA
(Moderna)
11 MoEdema, AKIPosNormal1.7 g/L22 g/L14.0 mg/dLPosPSL, amlodipine, clonidine, labetalol, RTX1 MoR
Renal function normalized
COVID-19 symptoms 6 Mo ago and edema 4 Mo ago[16]
468/MGermanyUNmRNA
(Pfizer-BioNTech)
17 dEdema, AKIPosUNPCR:
19.0 g/g
29 g/LUNUNIrbesartan4 MoNR
PCR: 14.4 g/g eGFR: decreased
-[17]
PCR:
14.0 g/g
UNUNRTX2 MoR
PCR: 6.0 g/g
eGFR: elevated
542/FUNUNAdenovirus vector
(AstraZeneca)
12 WEdema, foamy urine, hair lossUNNormalUN16 g/L0.8 mg/dLNegGC, HCQ, MMF,
diuretics
3 WR
Edema improved
ANA: 1:1280
COVID-19
antibody:32.8 U/mL
668/MWhiteUNAdenovirus
vector
(Johnson & Johnson)
1<4 WAKI, CKD,
NS
PosUN0.6 g/d32 g/L3.3 mg/dLUNDiuretics3 WR
UPRO: 0.4 g/d
Scr: 2.7 mg/dL
-[19]
757/FChinaNoInactivated
(Sinovac Life Sciences)
11 dEdemaPosNormal1.6 g/d29 g/L0.4 mg/dL48 RU/mLLosartan4 WR
Edema improved
-[20]
850/FWhiteUNmRNA
(Pfizer-BioNTech)
24 WJoint pain,
proteinuria
UN0.8 mg/dL6.5 g/d35 g/L0.7 mg/dLNegConservative2 MoR
UPRO: 0.4 g/d
ALB: 43 g/L
NELL-1-related,
hematuria
[21]
982/FItalyNomRNA
(Pfizer-BioNTech)
288 dNSUNUNUNUNUNNegGCUNUN-[22]
1067/FItalyNomRNA
(Pfizer-BioNTech)
289 dNSUNUNUNUNUNNegRTXUNUN-[22]
1182/MItalyNomRNA
(Pfizer-BioNTech)
229 dNSUNUNUNUNUNPosRTXUNUN-[22]
1280/MUNCKDUN2UNProteinuria,
hematuria
PosUNUNUNUNUNGC, RTX, CTXUNUNElevated p-ANCA,
c-ANCA and MPO,
positive of SSA-Ab
[23]
1380/FItalyTP, HT, IH, AVB,
hypothyroidism
mRNA
(Pfizer-BioNTech)
22 WEdema, AKIUN1.4 mg/dL10.0 g/d20 g/L2.0 mg/dLPosSteroids, CTX6 MoR
UPRO: 0.7 g/d, ALB: 40 g/L
FSGS, previously used steroids,
hematuria
[24]
1452/FItalyUNmRNA
(Pfizer-BioNTech)
249 dAsthma,
proteinuria
UNNormal3.4 g/dUN0.6 mg/dLUNRASB41 dR-[25]
15UNUNUNmRNA
(UN)
UN<3 WUNPosUNPCR:
4.5–7.6 g/g
36 g/L0.5–1.2mg/dLUNRASBUNUN-[26]
1654/MAsiaUNmRNA
(Moderna)
21 dNSNegUN3+34 g/L1.3 mg/dLUNSteroid, RTX8 WNR
UPRO: 3.5 g/d
Scr: 0.9 mg/dL
ANA: Pos,
ANCA: Pos,
hematuria
[19]
1747/MAsiaUNmRNA
(Moderna)
26 dNSNegUN2.7 g/d23 g/L0.7 mg/dLUNNo2 WR
UPRO: 2.7 g/d
Hematuria[19]
1822/MAustraliaEczema,
epilepsy
mRNA
(Pfizer-BioNTech)
21 MoEdema, lethargyPosNormalACR: 700.4 mg/mmol8 g/L0.7 mg/dL118 RU/mLPerindopril,
frusemide
anticoagulation
3 MoNR
PCR: 1.1 g/mmol
edema: worsened
-[27]
RTX2 MoR
PCR: 0.4 g/mmol
1932/MIndiaUNAdenovirus vector
(AstraZeneca)
UN14 dNS, thrombosisUNUNUNUNUNUNConservativeUNRCOVID-19
antibody:2334 U/mL
[28]
2047/MIndiaUNAdenovirus vector
(AstraZeneca)
UN11 dProteinuriaUNUNUNUNUNUNARBUNRCOVID-19
antibody:1500 U/mL
[28]
Relapsed or Worsening MN
177/FAmericaMNmRNA
(Pfizer-BioNTech)
14 WEdemaUN0.8 mg/dLPCR:
12.5 g/10 mmol
22 g/L0.7 mg/dL83 RU/mLTAC2 MoR-[29]
256/MAmericaMNmRNA
(Pfizer-BioNTech)
12 WEdema,
fatigue
UN1.5 mg/dL3.4 g/d32 g/L1.5 mg/dL30 RU/mLConservative7 MoNRRTX after 7 Mo, SARS-CoV-2
infected previously
[29]
365/FUNSystemic sarcoidosis, MNAdenovirus
vector
(Johnson& Johnson)
15 MoJoint, skin,
respiratory
symptoms, AKI
PosUNPCR:
3.4 g/g
UN1.7 mg/dLPosPSLUNR
PCR: 1.8 g/g
-[30]
467/MMalaysiaMNmRNA
(Pfizer-BioNTech)
12 WProteinuriaUNUN5.3 g/dUNUN42 RU/mLConservative1 MoR
UPRO: 1.6 g/d
CTX, steroids
previously
[31]
566/FTurkeyHT, DM, HLD, MNInactivated
(Sinovac Life Sciences)
12 WEdema, AKIUNNormalPCR:
9.4 mg/mg
26 g/L2.8 mg/dL121 RU/mLUNUNUNPreviously on
steroids and CsA but off 7 Y
[32]
648/MAmericaMNmRNA
(Pfizer-BioNTech)
12 WEdemaUN0.9 mg/dLUN25 g/L1.3 mg/dL155 RU/mLRTX, CTX, PSL3 MoRRTX and TAC were using during
vaccination
[29]
739/MWhiteMNmRNA
(Pfizer-BioNTech)
21 WEdemaPos0.9 mg/dL8.7 g/d20 g/L1.1 mg/dLUNTAC1 MoR
UPRO: 5.7 g/d
ALB: 29 g/L
Hematuria[21]
870/MWhiteMNmRNA
(Moderna)
24 WEdemaPos1.7 mg/dL16.6 g/d27 g/L2.1 mg/dLUNObinutuzumabUNUN-[21]
980/MAmericaMNmRNA
(Pfizer-BioNTech)
24 WEdemaUN1.2 mg/dL5.0 g/d26 g/L1.3 mg/dL916 RU/mLRTX4 MoNR-[29]
1060/MAmericaMNmRNA
(Pfizer-BioNTech)
26 WEdema, dry mouth, skin rash, AKIUN1.4 mg/dL5.0 g/d17 g/L1.9 mg/dL27 RU/mLRTX, CTX, PSL3 MoRTAC was using
during vaccination
[29]
1178/MAmericaMNmRNA
(Pfizer-BioNTech)
21 WEdema, HTUN1.6 mg/dLPCR: 4.9 g/10mmol34 g/L1.9 mg/dLNegPSL1 MoR-[29]
1248/MAmericaMNmRNA
(Pfizer-BioNTech)
23 WEdemaUN1.4 mg/dLPCR: 1.7 g/10mmol31 g/L1.4 mg/dL204 RU/mLConservative6 MoNRRTX, CTX and PSL after 6 Mo[29]
1362/FUNMetastatic breast Ca, HT, HLD, MNmRNA
(Moderna)
22 WEdema, dyspnea, proteinuria, AKIPosUN11.2 g/dUN1.6 mg/dL787 RU/mLLisinopril,
furosemide, RTX
UNR
PCR: 8.7 mg/g
-[33]
1484/MAmericaMNmRNA
(Pfizer-BioNTech)
210 WEdema,
dyspnea
UN1.5 mg/dL3.0 g/d33 g/L1.5 mg/dLPosTAC, PSL, RTX4 MoRSARS-CoV-2 infected previously, PSL and TAC were using
during vaccination
[29]
1539/MAmericaMNmRNA
(Pfizer-BioNTech)
24 WFatigueUN1.2 mg/dLPCR: 3.7 g/10mmol18 g/L1.4 mg/dL40 RU/mLRTX, CTX, PSL2 MoR-[29]
1675/MAmericaMNmRNA
(Pfizer-BioNTech)
22 WEdema,
fatigue
UN0.8 mg/dL8.0 g/d21 g/L0.9 mg/dL90 RU/mLRTX, CTX, PSLUNUN-[29]
1758/MAmericaMNmRNA
(Pfizer-BioNTech)
23 WEdemaUN1.0 mg/dL8.0 g/d24 g/L1.0 mg/dL22 RU/mLTAC2 MoRSARS-CoV-2 infected previously[29]
Abbreviations: AKI, acute kidney injury; ALB, albumin; ANA, antinuclear antibody; ANCA, antineutrophil cytoplasmic antibody; ARB, angiotensin receptor blocker; AVB, atrial ventricular block; Ca, cancer; CKD, chronic kidney disease; CsA, cyclosporin A; CTX, cyclophosphamide; d, day; DM, diabetes mellitus; eGFR, estimated glomerular filtration rate; F, female; FSGS, focal segmental glomerular sclerosis; FU, follow-up; GC, glucocorticoid; HCQ, hydroxychloroquine; HLD, hyperlipidemia; HT, hypertension; IH, ischaemic heart; M, male; MMF, mycophenolate mofetil; MN, membranous nephropathy; Mo, month; MPO, myeloperoxidase; NA, not applicable; Neg, negative; NELL-1, neural epidermal growth factor-like 1; NR, no response; NS, nephrotic syndrome; PCR, protein–creatinine ratio; PLA2R, phospholipase A2 receptor; PLA2R-Ab, phospholipase A2 receptor-antibody; Pos, positive; PSL, prednisone; R, response; RASB, renin-angiotensin system blockade; RTX, Rituximab; Scr, serum creatinine; SSA, Sjogren syndrome antigen A; TAC, tacrolimus; THSD7A-Ab, thrombospondin type-1 domain-containing 7A-antibody; TP, thrombocytopenic purpura; UN, unknown; UPRO, urine protein; W, week; Y, year.
Table 3. Clinical characteristics of MN post COVID-19 vaccination.
Table 3. Clinical characteristics of MN post COVID-19 vaccination.
CharacteristicsNew-Onset (n = 20) Relapsed or Worsening
(n = 17)
Total
(n = 37)
p
Age (years)57 (22–82)65 (39–84)63.5 (22–84)0.657
Male sex, n (%)12 (60.0)13 (76.5)25 (67.6)0.393
Medical history, n (%)-
Hypertension3 (15.0)2 (11.8)5 (13.5)-
Diabetes mellitus0 (0.0)1 (5.9)1 (2.7)-
Autoimmune disease0 (0.0)1 (5.9)1 (2.7)-
Vaccine type, n (%)0.672
mRNA15 (71.4) *15 (88.2)30 (78.9) *-
Pfizer-BioNTech10 (66.7)13 (86.7)23 (76.7)-
Moderna4 (26.7)2 (13.3)6 (20.0)-
Inactivated1 (4.8)1 (5.9)2 (5.3)-
Sinovac Life Sciences1 (100.0)1 (100.0)2 (100.0)-
Adenovirus vector4 (19.0)1 (5.9)5 (13.2)-
AstraZeneca3 (75.0)0 (0.0)3 (60.0)-
Johnson & Johnson1 (25.0)1 (100.0)2 (40.0)-
Unknown1 (4.8)0 (0.0)1 (2.6)-
Which dose, n (%)0.322
First dose7 (35.0)6 (35.3)13 (35.1)-
Second dose10 (50.0)11 (64.7)21 (56.8)-
Unknown3 (15.0)0 (0.0)3 (8.1)-
Onset time after vaccination, n (%)0.713
No more than 2 weeks10 (50.0)8 (47.1)18 (48.6)-
2 weeks-4 weeks4 (20.0)6 (35.3)10 (27.0)-
Beyond 4 weeks5 (25.0)3 (17.6)8 (21.6)-
Unknown1 (5.0)0 (0.0)1 (2.7)-
Symptoms, n (%)-
Edema14 (70.0)14 (82.4)28 (75.7)-
Proteinuria12 (60.0)2 (11.8)14 (37.8)-
Acute kidney injury6 (30.0)4 (23.5)10 (27.0)-
PLA2R stanning, n (%) **0.607
Positive7 (70.0)4 (100.0)11 (78.6)-
Negative3 (30.0)0 (0.0)3 (21.4)-
PLA2R-Ab, n (%) **0.065
Positive6 (54.5)14 (93.3)20 (76.9)-
Negative5 (45.5)1 (6.7)6 (23.1)-
Treatment, n (%)0.117
Immunosuppressive therapy11 (55.0)13 (76.5)24 (64.9)-
Conservative medication9 (45.0)3 (17.6)12 (32.4)-
Unknown0 (0.0)1 (5.9)1 (2.7)-
Outcome, n (%)0.795
Response13 (65.0)11 (64.7)24 (64.9)-
Not response2 (10.0)3 (17.6)5 (13.5)-
Unknown5 (25.0)3 (17.6)8 (21.6)-
* Data of one case was not available. ** In the population with known information, those without available data were not included.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ma, Q.; Li, X.; Xu, G. New-Onset and Relapsed Membranous Nephropathy post SARS-CoV-2 and COVID-19 Vaccination. Viruses 2022, 14, 2143. https://doi.org/10.3390/v14102143

AMA Style

Ma Q, Li X, Xu G. New-Onset and Relapsed Membranous Nephropathy post SARS-CoV-2 and COVID-19 Vaccination. Viruses. 2022; 14(10):2143. https://doi.org/10.3390/v14102143

Chicago/Turabian Style

Ma, Qiqi, Xiang Li, and Gaosi Xu. 2022. "New-Onset and Relapsed Membranous Nephropathy post SARS-CoV-2 and COVID-19 Vaccination" Viruses 14, no. 10: 2143. https://doi.org/10.3390/v14102143

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop