1
|
Kobayashi D, Yoshino J, Hanada M, Ohba M, Oka T, Itoga K, Niino D, Kanda T. A case of de novo glomerulonephritis following COVID-19 in a patient with preexistent IgA vasculitis. CEN Case Rep 2025; 14:236-241. [PMID: 39397227 PMCID: PMC11958849 DOI: 10.1007/s13730-024-00940-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
During the unprecedented COVID-19 outbreak, new-onset or relapsing glomerulonephritis, such as ANCA-associated glomerulonephritis and Immunoglobulin A (IgA) nephropathy, following COVID-19 has been reported. However, to date, the association of COVID-19 with preexistent IgA vasculitis (IgAV) remains unclear. Here, we present the case of a 20-something old Japanese woman with preexistent IgAV who newly developed glomerulonephritis following COVID-19. At the diagnosis of IgAV, she had cutaneous purpura, joint pains, and gastrointestinal symptoms, but no signs of kidney involvement. Three months ago, she was tested positive for COVID-19 and subsequently developed hematuria and proteinuria. She was then admitted to our hospital and renal biopsy showed glomerular mesangial expansion and hypercellularity and cellular and fibrocellular crescents, accompanied by diffuse IgA and C3 deposits. With the diagnosis of de novo IgAV nephritis, the patient was treated with intravenous methylprednisolone followed by oral prednisolone. She had favorable responses to this treatment and has achieved and maintained the remission of hematuria and proteinuria after initiation of glucocorticoid therapy. Our case highlights that immune response to SARS-CoV-2 infection could trigger the onset of glomerulonephritis in the IgAV patients who have no renal involvement.
Collapse
Affiliation(s)
- Daigo Kobayashi
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Jun Yoshino
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.
- The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Shimane, Japan.
| | - Maki Hanada
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Masafumi Ohba
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Tomohiro Oka
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Kenichi Itoga
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Daisuke Niino
- Department of Functional Pathology, Faculty of Medicine, Shimane University, Izumo, Shimane, Japan
| | - Takeshi Kanda
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
- The Center for Integrated Kidney Research and Advance (IKRA), Faculty of Medicine, Shimane University, Izumo, Shimane, Japan
| |
Collapse
|
2
|
Lim HX, Khalid K, Abdullah ADI, Lee LH, Raja Ali RA. Subphenotypes of Long COVID and the clinical applications of probiotics. Biomed Pharmacother 2025; 183:117855. [PMID: 39862702 DOI: 10.1016/j.biopha.2025.117855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
As the number of infections and deaths attributable to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection continues to rise, it is now becoming apparent that the health impacts of the Coronavirus disease (COVID-19) may not be limited to infection and the subsequent resolution of symptoms. Reports have shown that patients with SARS-CoV-2 infection may experience multiple symptoms across different organ systems that are associated with adverse health outcomes and develop new cardiac, renal, respiratory, musculoskeletal, and nervous conditions, a condition known as Long COVID or the post-acute sequelae of COVID-19 (PASC). This review provides insights into distinct subphenotypes of Long COVID and identifies microbiota dysbiosis as a common theme and crucial target for future therapies. Another important finding is that Long COVID is associated with prolonged and increased inflammation, potentially attributable to immune system dysfunction. A promising solution lies in the potential of probiotics to mitigate Long COVID symptoms by restoring gut microbiota balance and modulating the immune response. By evaluating the current clinical development landscape of the use of probiotics to treat Long COVID symptoms, this paper provides recommendations for future research by stressing the need to understand the modulation of bacterium strains followed by probiotic therapy to understand the association of microbiota dysbiosis with Long COVID symptoms. This will facilitate the development of effective probiotic formulations that could serve as reliable therapies against Long COVID.
Collapse
Affiliation(s)
- Hui Xuan Lim
- Sunway Microbiome Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia.
| | - Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia.
| | | | - Learn-Han Lee
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham, Ningbo 315000, China
| | - Raja Affendi Raja Ali
- School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia.
| |
Collapse
|
3
|
Qi L, Deep A, Fox J, Yii M, Rahman M, Myint M, Myat H, Thet Z. A scoping review on adult patients with de novo glomerular diseases following COVID-19 infection or vaccine. Int Urol Nephrol 2025; 57:447-462. [PMID: 39225763 PMCID: PMC11772384 DOI: 10.1007/s11255-024-04189-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND There are increasing reports of glomerular disease (GD) following COVID-19 infection and vaccination. Current evidence on the possible link between COVID-19 infection or vaccination and GD is conflicting. OBJECTIVE The present study undertakes a scoping review of research to describe the relationship between COVID-19 infection and vaccination with GD and the common management strategies and overall outcomes of the disease to identify knowledge gaps and guide further research. ELIGIBILITY CRITERIA All original research studies published in English until 5th September 2022 were considered for inclusion in the review. Exclusion criteria were animal studies, autopsy studies, and data involving patients who were paediatric patients (< 16 years), were transplant recipients, had a recurrence of glomerular disease, had concomitant cancer or non-COVID-19 infection which may cause glomerular disease, or did not receive a renal biopsy. SOURCES OF EVIDENCE The five electronic databases searched were MEDLINE, PubMed, Scopus, EMBASE, and Cochrane. METHODS Two separate search strings related to COVID-19, and glomerular disease were combined using the Boolean operator 'AND'. Filters were used to limit publications to original research studies published in English. Search results from each database were imported into Covidence software ( www.covidence.org ) and used for de-duplication, article screening, and data extraction. Descriptive analyses were used to summarise demographics, diagnoses, and treatment outcomes. RESULTS After removing duplicates, 6853 titles and abstracts were screened. Of the 188 studies included, 106 studies described 341 patients with GD following COVID-19 infection and 82 described 146 patients with GD following a COVID-19 vaccination. IgA nephropathy was the most common GD pathology reported following COVID-19 vaccination with GD most common following mRNA vaccines. Collapsing focal segmental glomerulosclerosis was the most common GD following COVID-19 infection. Immunosuppressive treatment of GD was more common in the vaccine cohort than in the infection cohort. CONCLUSION Despite the significant number of COVID-19 infections and vaccinations around the world, our understanding of GD associated with COVID-19 infection and vaccination remains poor, and more research is needed to understand the possible relationship better.
Collapse
Affiliation(s)
- Liam Qi
- Department of Medicine, Prince Charles Hospital, Metro North Hospital and Health Service, Chermside, QLD, Australia
- The University of Queensland, Faculty of Medicine, Brisbane, QLD, Australia
| | - Aman Deep
- Department of Nephrology, Central Queensland Hospital and Health Service, Rockhampton, QLD, Australia
| | - Jordan Fox
- The University of Queensland Rural Clinical School, Rockhampton, QLD, Australia
| | - Mark Yii
- Department of Medicine, Flinders Medical Centre, South Adelaide Local Health Network, Bedford Park, SA, Australia
| | - Muhammad Rahman
- The University of Queensland Rural Clinical School, Rockhampton, QLD, Australia
| | - Mar Myint
- K & K Kidney Health, Rockhampton, QLD, Australia
| | - Htoo Myat
- Department of Medicine, Canberra Hospital, Canberra Hospital and Health Service, Garran, ACT, Australia
| | - Zaw Thet
- Department of Nephrology, Central Queensland Hospital and Health Service, Rockhampton, QLD, Australia.
- K & K Kidney Health, Rockhampton, QLD, Australia.
- The University of Queensland Rural Clinical School, Rockhampton, QLD, Australia.
- School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
4
|
Wang G, Yang L, Xu X, Guo W, Sun L, Wang Y, Cheng W, Ye N, Kong L, Zhao X, Cheng H. SARS-CoV-2 Protein Deposition Enhances Renal Complement Activation and Aggravates Kidney Injury in Membranous Nephropathy After COVID-19. Kidney Int Rep 2024; 9:3145-3155. [PMID: 39534192 PMCID: PMC11551107 DOI: 10.1016/j.ekir.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction COVID-19 has been reported to be associated with the occurrence and recurrence of membranous nephropathy (MN). The clinicopathological characteristics and complement system activation of MN after COVID-19 are unclear. Methods A total of 38 patients with biopsy-proven MN who developed new-onset proteinuria after COVID-19 were enrolled in this study. One hundred patients with primary MN diagnosed before the COVID-19 pandemic were the control. Renal immunohistochemical staining for SARS-CoV-2 nucleocapsid protein was performed in 38 patients with MN after COVID-19. Serum membrane attack complex (MAC) was detected by enzyme-linked immunosorbent assay. Glomerular staining for the complement proteins in different pathways were detected by immunohistochemistry. Results Thirteen of 38 patients had positive staining for SARS-CoV-2 nucleocapsid protein. Compared with the control patients, the clinical manifestations were more severe in patients after COVID-19. Patients with positive SARS-CoV-2 staining had a higher proportion of nephrotic syndrome, lower level of serum albumin, and greater severity of renal interstitial fibrosis than those of patients with negative SARS-CoV-2 staining. Serum MAC level and renal MAC staining intensity of MN after COVID-19 were significantly higher than those of the control patients. MAC expression in MN patients with positive SARS-CoV-2 staining was stronger than that in both control patients and MN after COVID-19 with negative SARS-CoV-2 staining. The expression trend of factor H was consistent with that of MAC. Conclusion Excessive activation of the complement system aggravated symptoms in MN after COVID-19. Therapeutic strategy targeting the complement system may need to be considered.
Collapse
Affiliation(s)
- Guoqin Wang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lei Yang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyi Xu
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Weiyi Guo
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Sun
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanyan Wang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wenrong Cheng
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Nan Ye
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lingqiang Kong
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyi Zhao
- Division of Nephrology, Affiliated Hospital of Chifeng University, Neimenggu, China
| | - Hong Cheng
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Fu N, Yuan S, Yang G, Li H, Wang T. Concurrent glomerular PCDH7 deposits in PLA2R-associated membranous nephropathy. CEN Case Rep 2024; 13:297-301. [PMID: 38145434 PMCID: PMC11294502 DOI: 10.1007/s13730-023-00842-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
The understanding of membranous nephropathy (MN) has undergone impressive advancements in the last 5 years, particularly due to identification of novel antigenic targets. M-type phospholipase A2 receptor (PLA2R) and thrombospondin type 1 domain-containing 7A (THSD7A) account for approximately 70% and 1-5% of the target antigens in primary MN, respectively. Recently, more novel/putative antigens have been identified in the remaining cases of MN that include exostosin 1/exostosin 2 (EXT1/EXT2), neural epidermal growth factor-like 1 protein (NELL-1), semaphorin 3B (SEMA3B) and protocadherin 7 (PCDH7). However, comparatively little is known about the PCDH7 among these novel antigens. As such, we herein described a unique case of positive glomerular PCDH7 deposits in PLA2R-associated MN, which may offer a deeper insight into the role of PCDH7 in MN and improve our understanding of glomerular diseases in the post-COVID era, particularly with the emerging variants.
Collapse
Affiliation(s)
- NaNa Fu
- Department of Nephrology, ShouGuang Chinese Medicine Hospital, No.3353 ShengCheng Street, ShouGuang, 262700, People's Republic of China
| | - Shuang Yuan
- Graduate School of HeBei Medical University, No.361 East ZhongShan Boulevard, Shijiazhuang, 050011, People's Republic of China
| | - Guang Yang
- TaiYuan KingMed Center for Clinical Laboratory, No.2 LongSheng Street, Taiyuan, 030000, People's Republic of China
| | - Hang Li
- Department of Nephrology, Peking Union Medical College Hospital, No.1 ShuaiFuYuan, Beijing, 100010, People's Republic of China
| | - Tao Wang
- Department of Nephrology, The First Hospital of HeBei Medical University, No.89 DongGang Road, Shijiazhuang, 050030, People's Republic of China.
| |
Collapse
|
6
|
Li C, Zhang C, Chen J, Chen Y, Ying Z, Hu Y, Song H, Fu P, Zeng X. The Time-Varying Impact of COVID-19 on the Acute Kidney Disorders: A Historical Matched Cohort Study and Mendelian Randomization Analysis. HEALTH DATA SCIENCE 2024; 4:0159. [PMID: 39011273 PMCID: PMC11246837 DOI: 10.34133/hds.0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024]
Abstract
Background: This study aimed to explore the time-varying impact of COVID-19 on acute kidney disorders, including acute kidney injury and other acute kidney diseases. Methods: From the UK Biobank, 10,121 participants with COVID-19 were matched with up to 3 historically unexposed controls by age, sex, Townsend deprivation index, and the status of hospitalization or receiving critical care. We investigated the association between COVID-19 and incidence of acute kidney disorders, within the first 4 weeks after infection, using conditional and time-varying Cox proportional hazard regression. In addition, one-sample Mendelian randomization, utilizing the polygenic risk score for COVID-19 as an instrumental variable, was conducted to explore the potential causality of the association. Results: In the matched cohort study, we observed a significant association between COVID-19 and acute kidney disorders predominantly within the first 3 weeks. The impact of COVID-19 was time dependent, peaking in the second week (hazard ratio, 12.77; 95% confidence interval, 5.93 to 27.70) and decreasing by the fourth week (hazard ratio, 2.28; 95% confidence interval, 0.75 to 6.93). In subgroup analyses, only moderate to severe COVID-19 cases were associated with acute worsening of renal function in a time-dependent pattern. One-sample Mendelian randomization analyses further showed that COVID-19 might exert a "short-term" causal effect on the risk of acute kidney disorders, primarily confined to the first week after infection. Conclusions: The risk of acute kidney disorders following COVID-19 demonstrates a time-varying pattern. Hazard effects were observed only in patients with moderate or severe but not mild COVID-19.
Collapse
Affiliation(s)
- Chunyang Li
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
| | - Chao Zhang
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
| | - Jie Chen
- Department of Core Laboratory, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yilong Chen
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
| | - Zhiye Ying
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
| | - Yao Hu
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
| | - Huan Song
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
- Centre of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Ping Fu
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoxi Zeng
- Division of Nephrology, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu 610065, China
| |
Collapse
|
7
|
Fisher M, Ross M, DiFranza L, Reidy K. An Update on Viral Infection-Associated Collapsing Glomerulopathy. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:317-325. [PMID: 39084757 PMCID: PMC11296492 DOI: 10.1053/j.akdh.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/14/2023] [Accepted: 12/14/2023] [Indexed: 08/02/2024]
Abstract
The COVID-19 era has been a reminder to clinicians around the world of the important role that viral infections play in promoting glomerular disease. Several viral infections including human immunodeficiency virus (HIV), severe acute respiratory syndrome coronavirus 2, Epstein-Barr virus, cytomegalovirus, and parvovirus B19 can cause podocyte injury and present with a collapsing glomerulopathy (CG) variant of focal segmental glomerulosclerosis or minimal change disease. CG associated with COVID-19 has been termed COVID-19-associated nephropathy due to its striking resemblance to HIV-associated nephropathy. Host susceptibility is a major determinant of viral infection-associated CG, and the presence of two APOL1 risk variants explains most of the racial predilection to viral-associated CG observed in individuals of African ancestry. Interactions between APOL1 risk variants, viral genes, and the systemic inflammatory response to viral infection all contribute to kidney injury. This review will summarize our current knowledge of viral infection-associated CG, focusing primarily on the clinical presentation, histological features, mechanisms, and disease course of HIV-associated nephropathy and COVID-19-associated nephropathy.
Collapse
Affiliation(s)
- Molly Fisher
- Division of Nephrology, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY.
| | - Michael Ross
- Division of Nephrology, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Lanny DiFranza
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY
| | - Kimberly Reidy
- Division of Pediatric Nephrology, The Children's Hospital at Montefiore/Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
8
|
Hui M, Shi SF, Zhou XJ, Liu LJ, Lv JC, Zhang H. The impact of mild and moderate COVID-19 infection on the progression of kidney dysfunction in patients with IgA nephropathy. Clin Kidney J 2024; 17:sfae157. [PMID: 38979107 PMCID: PMC11229029 DOI: 10.1093/ckj/sfae157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Indexed: 07/10/2024] Open
Abstract
Background Previous research indicates that coronavirus disease 2019 (COVID-19) infection may have a role in triggering immunoglobulin A (IgA) nephropathy. However, limited research has explored the clinical implications of COVID-19 infection in individuals already diagnosed with IgA nephropathy. This study aimed to determine whether COVID-19 infection independently affects the subsequent trajectory of kidney function in IgA nephropathy patients. Methods This was a single-center cohort study. The study included 199 patients diagnosed with IgA nephropathy. The COVID-19 infection status was determined using a combined method: a questionnaire and the Health Code application, both administered at the end of 2022 in northern China. Kidney function trajectory was assessed by the estimated glomerular filtration rate (eGFR), calculated based on serum creatinine levels measured during follow-up outpatient visits. The primary endpoint of interest was the eGFR trajectory. Results Out of the 199 participants, 75% (n = 181) reported a confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, determined through antigen or polymerase chain reaction tests, accounting for 79% (n = 143) of the infected patients. A significant majority (98%) experienced mild to moderate symptoms. Over a median follow-up period of 10.7 months post-COVID-19 infection, notable clinical events included gross hematuria in 30 patients (16.6%), which normalized within an average of 3 days. Additionally, a 2-fold increase in proteinuria or progression to the nephrotic range was observed in 10 individuals (5.5%). No cases of acute kidney injury were noted. COVID-19 exposure was associated with an absolute change in eGFR of 2.98 mL/min/1.73 m2 per month (95% confidence interval 0.46 to 5.50). However, in a fully adjusted model, the estimated changes in eGFR slope post-COVID-19 were -0.39 mL/min/1.73 m2 per month (95% confidence interval -0.83 to 0.06, P = .088) which included the possibility of no significant effect. Notably, a higher rate of kidney function decline was primarily observed in patients with a baseline eGFR <45 mL/min/1.73 m2 [-0.56 mL/min/1.73 m2 (-1.11 to -0.01), P = .048]. In the cohort, there were few instances of severe COVID-19 cases. The absence of long-term follow-up outcomes was observed. Conclusions Overall, mild to moderate COVID-19 infection does not appear to significantly exacerbate the subsequent decline in kidney function among IgA nephropathy patients, particularly in those with preserved baseline kidney function.
Collapse
Affiliation(s)
- Miao Hui
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, National Health Commission of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Su-Fang Shi
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, National Health Commission of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xu-Jie Zhou
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, National Health Commission of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li-Jun Liu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, National Health Commission of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ji-Cheng Lv
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, National Health Commission of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, National Health Commission of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Tabachnikov O, Skorecki K, Kruzel-Davila E. APOL1 nephropathy - a population genetics success story. Curr Opin Nephrol Hypertens 2024; 33:447-455. [PMID: 38415700 PMCID: PMC11139250 DOI: 10.1097/mnh.0000000000000977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
PURPOSE OF REVIEW More than a decade ago, apolipoprotein L1 ( APOL1 ) risk alleles designated G1 and G2, were discovered to be causally associated with markedly increased risk for progressive kidney disease in individuals of recent African ancestry. Gratifying progress has been made during the intervening years, extending to the development and clinical testing of genomically precise small molecule therapy accompanied by emergence of RNA medicine platforms and clinical testing within just over a decade. RECENT FINDINGS Given the plethora of excellent prior review articles, we will focus on new findings regarding unresolved questions relating mechanism of cell injury with mode of inheritance, regulation and modulation of APOL1 activity, modifiers and triggers for APOL1 kidney risk penetrance, the pleiotropic spectrum of APOL1 related disease beyond the kidney - all within the context of relevance to therapeutic advances. SUMMARY Notwithstanding remaining controversies and uncertainties, promising genomically precise therapies targeted at APOL1 mRNA using antisense oligonucleotides (ASO), inhibitors of APOL1 expression, and small molecules that specifically bind and inhibit APOL1 cation flux are emerging, many already at the clinical trial stage. These therapies hold great promise for mitigating APOL1 kidney injury and possibly other systemic phenotypes as well. A challenge will be to develop guidelines for appropriate use in susceptible individuals who will derive the greatest benefit.
Collapse
Affiliation(s)
- Orly Tabachnikov
- Department of Nephrology, Rambam Healthcare Campus, Haifa, Israel
| | - Karl Skorecki
- Department of Nephrology, Rambam Healthcare Campus, Haifa, Israel
- Departments of Genetics and Developmental Biology and Rappaport Faculty of Medicine and Research Institute, Technion—Israel Institute of Technology, Haifa, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Etty Kruzel-Davila
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Department of Nephrology, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
10
|
Sakai H, Kamuro H, Tokunoh N, Izawa T, Tamiya S, Yamamoto A, Tanaka S, Okuzaki D, Ono C, Matsuura Y, Okada Y, Yoshioka Y, Fujio Y, Obana M. JAK inhibition during the early phase of SARS-CoV-2 infection worsens kidney injury by suppressing endogenous antiviral activity in mice. Am J Physiol Renal Physiol 2024; 326:F931-F941. [PMID: 38634132 PMCID: PMC11381010 DOI: 10.1152/ajprenal.00011.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) induces respiratory dysfunction as well as kidney injury. Although the kidney is considered a target organ of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and affected by the COVID-19-induced cytokine storm, the mechanisms of renal reaction in SARS-CoV-2 infection are unknown. In this study, a murine COVID-19 model was induced by nasal infection with mouse-adapted SARS-CoV-2 (MA10). MA10 infection induced body weight loss along with lung inflammation in mice 4 days after infection. Serum creatinine levels and the urinary albumin/creatinine ratio increased on day 4 after MA10 infection. Measurement of the urinary neutrophil gelatinase-associated lipocalin/creatinine ratio and hematoxylin and eosin staining revealed tubular damage in MA10-infected murine kidneys, indicating kidney injury in the murine COVID-19 model. Interferon (IFN)-γ and interleukin-6 upregulation in the sera of MA10-infected mice, along with the absence of MA10 in the kidneys, implied that the kidneys were affected by the MA10 infection-induced cytokine storm rather than by direct MA10 infection of the kidneys. RNA-sequencing analysis revealed that antiviral genes, such as the IFN/Janus kinase (JAK) pathway, were upregulated in MA10-infected kidneys. Upon administration of the JAK inhibitor baricitinib on days 1-3 after MA10 infection, an antiviral pathway was suppressed, and MA10 was detected more frequently in the kidneys. Notably, JAK inhibition upregulated the hypoxia response and exaggerated kidney injury. These results suggest that endogenous antiviral activity protects against SARS-CoV-2-induced kidney injury in the early phase of infection, providing valuable insights into the pathogenesis of COVID-19-associated nephropathy.NEW & NOTEWORTHY Patients frequently present with acute kidney injury or abnormal urinary findings after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here, we investigated how the kidneys respond during SARS-CoV-2 infection using a murine coronavirus disease 2019 (COVID-19) model and showed that Janus kinase-mediated endogenous antiviral activity protects against kidney injury in the early phase of SARS-CoV-2 infection. These findings provide valuable insights into the renal pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Hibiki Sakai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyasu Kamuro
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Nagisa Tokunoh
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Research Foundation for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Osaka Metropolitan University Graduate School of Veterinary Science, Osaka, Japan
| | - Shigeyuki Tamiya
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Department of Microbiology and Immunology, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Ayaha Yamamoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Chikako Ono
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Yasuo Yoshioka
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Research Foundation for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
- Radioisotope Research Center, Institute for Radiation Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
11
|
Diebold M, Enzler-Tschudy A, Helmchen BM, Hopfer H, Kim MJ, Moll S, Nanchen G, Rotman S, Seeger H, Kistler AD. Incidence of Common Glomerular Diseases Other Than Collapsing Glomerulopathy is Not Increased After SARS-CoV-2 Infection. Kidney Int Rep 2024; 9:1122-1126. [PMID: 38765571 PMCID: PMC11101795 DOI: 10.1016/j.ekir.2024.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 05/22/2024] Open
Affiliation(s)
- Matthias Diebold
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Birgit M. Helmchen
- Institute of Pathology and Molecular Pathology, University of Zurich, Zurich, Switzerland
| | - Helmut Hopfer
- Department of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Min Jeong Kim
- Division of Nephrology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Solange Moll
- Service of clinical Pathology, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | - Giliane Nanchen
- Service of Nephrology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Samuel Rotman
- Service of Clinical Pathology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Harald Seeger
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
- Institute for Nephrology and Dialysis, Cantonal Hospital Baden, Baden, Switzerland
| | - Andreas D. Kistler
- Department of Medicine, Cantonal Hospital Frauenfeld, Spital Thurgau AG, Frauenfeld, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
de Sousa MV. Post-Transplant Glomerulonephritis: Challenges and Solutions. Int J Nephrol Renovasc Dis 2024; 17:81-90. [PMID: 38495741 PMCID: PMC10944656 DOI: 10.2147/ijnrd.s391779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/12/2024] [Indexed: 03/19/2024] Open
Abstract
Glomeruli can be damaged in several conditions after kidney transplantation, with a potential impact on the graft function and survival. Primary glomerulonephritis, a group of glomerular immunological damage that results in variable histological patterns and clinical phenotypes, can occur in kidney transplant recipients as a recurrent or de novo condition. Specific immunologic conditions associated with kidney transplantation, such as acute rejection episodes, can act as an additional trigger after transplantation, impacting the incidence of these glomerulopathies. The post-transplant GN recurrence ranges from 3% to 15%, varying according to the GN subtype and post-transplant time, mainly occurring after 3-5 years of kidney transplantation. Advances in the knowledge of glomerulonephritis pathophysiology have provided new approaches to pre-transplant risk evaluation and post-transplant monitoring. Glomeruli can be affected by several systemic viral infections, such as human immunodeficiency virus (HIV), hepatitis C virus (HCV), hepatitis B virus (HBV), severe acute respiratory syndrome coronavirus 2 (SARS-COV-2), cytomegalovirus (CMV), and BK virus. The diagnosis of these infections, as well as the identification of possible complications associated with them, are important to minimize the negative impacts of these conditions on kidney transplant recipients' outcomes.
Collapse
Affiliation(s)
- Marcos Vinicius de Sousa
- University of Campinas, School of Medical Sciences, Department of Internal Medicine, Division of Nephrology, Renal Transplant Unit, Transplant Research Laboratory, Campinas, SP, Brazil
| |
Collapse
|
13
|
Kim HW, Kim EH, Roh YH, Joo YS, Eom M, Kim HS, Kang MS, Jeong H, Lim BJ, Han SH, Jung M. Glomerulonephritis following COVID-19 infection or vaccination: a multicenter study in South Korea. Kidney Res Clin Pract 2024; 43:165-176. [PMID: 38600028 PMCID: PMC11016674 DOI: 10.23876/j.krcp.23.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/16/2023] [Accepted: 01/01/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Despite the widespread impact of the severe acute respiratory syndrome coronavirus 2 (coronavirus disease 2019, COVID-19) and vaccination in South Korea, our understanding of kidney diseases following these events remains limited. We aimed to address this gap by investigating the characteristics of glomerular diseases following the COVID-19 infection and vaccination in South Korea. METHODS Data from multiple centers were used to identify de novo glomerulonephritis (GN) cases with suspected onset following COVID-19 infection or vaccination. Retrospective surveys were used to determine the COVID-19-related histories of patients who were initially not implicated. Bayesian structural time series and autoregressive integrated moving average models were used to determine causality. RESULTS Glomerular diseases occurred shortly after the infection or vaccination. The most prevalent postinfection GN was podocytopathy (42.9%), comprising primary focal segmental glomerulosclerosis and minimal change disease, whereas postvaccination GN mainly included immunoglobulin A nephropathy (IgAN; 57.9%) and Henoch-Schönlein purpura nephritis (HSP; 15.8%). No patient progressed to end-stage kidney disease. Among the patients who were initially not implicated, nine patients with IgAN/HSP were recently vaccinated against COVID-19. The proportion of glomerular diseases changed during the pandemic in South Korea, with an increase in acute interstitial nephritis and a decrease in pauci-immune crescentic GN. CONCLUSION This study showed the characteristics of GNs following COVID-19 infection or vaccination in South Korea. Understanding these associations is crucial for developing effective patient management and vaccination strategies. Further investigation is required to fully comprehend COVID-19's impact on GN.
Collapse
Affiliation(s)
- Hyung Woo Kim
- Department of Internal Medicine, Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Hwa Kim
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yun Ho Roh
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Su Joo
- Division of Nephrology, Department of Internal Medicine, Yongin Severance Hospital, Yongin, Republic of Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Han Seong Kim
- Department of Pathology, Inje University Ilsan Paik Hospital, Goyang, Republic of Korea
| | - Mi Seon Kang
- Department of Pathology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - HoeIn Jeong
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Beom Jin Lim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minsun Jung
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Renal Pathology Study Group of Korean Society of Pathologists
- Department of Internal Medicine, Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, Yongin Severance Hospital, Yongin, Republic of Korea
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
- Department of Pathology, Inje University Ilsan Paik Hospital, Goyang, Republic of Korea
- Department of Pathology, Inje University Busan Paik Hospital, Busan, Republic of Korea
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
14
|
de Araújo EMC, Campos MAG, Sodré AM, de Holanda MI, Hagemann R, Teixeira Júnior AAL, Salgado Filho N, Neves PDMDM, Silva GEB. Tip Lesion Most Frequent FSGS Variant Related to COVID-19 Vaccine: Two Case Reports and Literature Review. Vaccines (Basel) 2024; 12:62. [PMID: 38250875 PMCID: PMC10821173 DOI: 10.3390/vaccines12010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Large-scale COVID-19 vaccination has been one of the most effective strategies to control the spread of the SARS-CoV-2 virus. However, several cases of glomerular injury related to the COVID-19 vaccine have been described in the literature. We report two cases of a tip lesion variant of focal segmental glomerulosclerosis (FSGS), which presented with significant proteinuria and improved after immunosuppression. In our literature review, the tip lesion variant of FSGS is currently the most frequent variant associated with vaccination against COVID-19. Prognosis is favorable and without significant alterations in the tubulointerstitial or vascular compartments. Adverse effects of vaccines need to be recognized early and will help us to understand the immune and pathological mechanisms of kidney damage.
Collapse
Affiliation(s)
- Emmy Marjorie Carvalho de Araújo
- Faculty of Medicine, Federal University of Maranhão, Gonçalves Dias Square, São Luís 65020-240, Brazil; (E.M.C.d.A.); (A.M.S.); (N.S.F.)
| | - Marcos Adriano Garcia Campos
- Clinical Hospital of Botucatu Medical School, São Paulo State University, Professor Mário Rubens Guimarães Montenegro Avenue, Botucatu 18618-687, Brazil;
| | - Andressa Monteiro Sodré
- Faculty of Medicine, Federal University of Maranhão, Gonçalves Dias Square, São Luís 65020-240, Brazil; (E.M.C.d.A.); (A.M.S.); (N.S.F.)
| | | | - Rodrigo Hagemann
- Clinical Hospital Complex, Federal University of Paraná, General Carneiro Street, Curitiba 80060-900, Brazil;
| | | | - Natalino Salgado Filho
- Faculty of Medicine, Federal University of Maranhão, Gonçalves Dias Square, São Luís 65020-240, Brazil; (E.M.C.d.A.); (A.M.S.); (N.S.F.)
| | | | - Gyl Eanes Barros Silva
- Faculty of Medicine, Federal University of Maranhão, Gonçalves Dias Square, São Luís 65020-240, Brazil; (E.M.C.d.A.); (A.M.S.); (N.S.F.)
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, Bandeirantes Avenue, Ribeirão Preto 14040-900, Brazil
| |
Collapse
|
15
|
Jung J, Lee JH. Clinical considerations and practical issues of kidney complications in children after COVID-19 infection or vaccination. Clin Exp Pediatr 2024; 67:35-36. [PMID: 37986565 PMCID: PMC10764664 DOI: 10.3345/cep.2023.01074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/19/2023] [Indexed: 11/22/2023] Open
Affiliation(s)
- Jiwon Jung
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Joo Hoon Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
16
|
Carvajal Abreu K, Loos S, Fischer L, Pape L, Wiech T, Kemper MJ, Tönshoff B, Oh J, Schild R. Case report: Early onset de novo FSGS in a child after kidney transplantation-a successful treatment. Front Pediatr 2023; 11:1280521. [PMID: 37830056 PMCID: PMC10566293 DOI: 10.3389/fped.2023.1280521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 09/14/2023] [Indexed: 10/14/2023] Open
Abstract
Background Early onset de novo focal segmental glomerular sclerosis (FSGS) in the kidney allograft in patients without FSGS in the native kidney is a rare disorder in children. It usually occurs mostly beyond the first year after kidney transplantation and often leads to graft loss. Standardized treatment protocols have not yet been established. Case description We describe a boy with early onset de novo FSGS in the transplanted kidney and non-selective glomerular proteinuria (maximum albumin-to-creatinine ratio of 3.8 g/g; normal range, ≤0.03 g/g creatinine). Manifestation occurred at 30 days posttransplant and was accompanied by a significant graft dysfunction (eGFR 61 ml/min per 1.73 m2). Treatment with 25 sessions of plasmapheresis over 14 weeks and three consecutive days of methylprednisolone pulse therapy (10 mg/kg per day) followed by oral prednisolone as rejection prophylaxis (3.73 mg/m2 per day) led to sustained remission of proteinuria (albumin-to-creatinine ratio of 0.028 g/g) and normalization of graft function (eGFR 92 ml/min per 1.73 m2) after 14 weeks. The follow-up period was 36 months. Conclusions This case underlines the efficacy of immunosuppressive and antibody eliminating therapy in early onset de novo FSGS after kidney transplantation.
Collapse
Affiliation(s)
- Karla Carvajal Abreu
- Department of Pediatric Nephrology, Pediatric Hepatology and Pediatric Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian Loos
- Department of Pediatric Nephrology, Pediatric Hepatology and Pediatric Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lutz Fischer
- Department of Hepatobiliary Surgery and Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Pape
- Department of Pediatrics II, University Hospital of Essen, University of Essen-Duisburg, Essen, Germany
| | - Thorsten Wiech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus J. Kemper
- Department of Pediatrics, Asklepios Klinik Nord-Heidberg, Hamburg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Jun Oh
- Department of Pediatric Nephrology, Pediatric Hepatology and Pediatric Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Raphael Schild
- Department of Pediatric Nephrology, Pediatric Hepatology and Pediatric Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|