1
|
Lee T, Ma L, Freedman BI. APOL1 testing in clinical practice and opportunities for new therapies. Curr Opin Nephrol Hypertens 2025:00041552-990000000-00232. [PMID: 40314119 DOI: 10.1097/mnh.0000000000001082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
PURPOSE OF REVIEW The spectrum of kidney diseases caused by variation in the apolipoprotein L1 (APOL1) gene was identified in 2010 among patients with recent African ancestry. In the United States, inheriting two APOL1 risk variants (high-risk genotypes) markedly increases risk for solidified glomerulosclerosis, focal segmental glomerulosclerosis, collapsing glomerulopathy, lupus nephritis, and sickle cell nephropathy. Kidneys from African American deceased donors with APOL1 high-risk genotypes also fail more rapidly after transplant. One risk variant increases nephropathy risk in Africa. This review focuses on novel therapies targeting APOL1 and the changing landscape of APOL1 genotyping in patients at risk for APOL1-mediated kidney disease (AMKD). RECENT FINDINGS Renin-angiotensin-aldosterone system blockade and sodium-glucose cotransporter 2 inhibitors slow nephropathy progression but are not curative. Medications directly targeting APOL1 mRNA and blocking APOL1 protein effects are undergoing clinical trials in AMKD, including APOL1 small molecule inhibitors, an APOL1 antisense oligonucleotide, and a Janus kinase (JAK) signaling inhibitor to reduce APOL1 expression. Early results are promising and provide hope for well tolerated and effective therapies. If successful, more patients will need to be considered for APOL1 genotyping, and our approach to diagnosing and treating chronic kidney disease in populations with recent African ancestry will change dramatically. SUMMARY Mechanisms of APOL1 risk variant nephrotoxicity remain unclear; nonetheless, specific therapies for AMKD show great promise and may improve understanding of disease processes. With ongoing clinical trials and the potential for effective AMKD treatments, more widespread APOL1 genotyping will likely be needed.
Collapse
Affiliation(s)
- Taewoo Lee
- Department of Internal Medicine, Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | | |
Collapse
|
2
|
Hung RKY, Costeira R, Chen J, Schlosser P, Grundner-Culemann F, Booth JW, Sharpe CC, Bramham K, Sun YV, Marconi VC, Teumer A, Winkler CA, Post FA, Bell JT. Epigenetic associations with kidney disease in individuals of African ancestry with APOL1 high-risk genotypes and HIV. Nephrol Dial Transplant 2025; 40:997-1006. [PMID: 39448372 PMCID: PMC12035534 DOI: 10.1093/ndt/gfae237] [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: 03/25/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Apolipoprotein L1 (APOL1) high-risk variants are major determinants of chronic kidney disease (CKD) in people of African ancestry. Previous studies have identified epigenetic changes in relation to kidney function and CKD, but not in individuals with APOL1 high-risk genotypes. We conducted an epigenome-wide analysis of CKD and estimated glomerular filtration rate (eGFR) in in people of African ancestry and APOL1 high-risk genotypes with HIV. METHODS DNA methylation profiles from peripheral blood mononuclear cells of 119 individuals with APOL1 high-risk genotypes (mean age 48 years, 49% female, median CD4 count 515 cells/mm3, 90% HIV-1 RNA <200 copies/mL, 23% with CKD) were obtained by Illumina MethylationEPIC BeadChip. Differential methylation analysis of CKD considered technical and biological covariates. We also assessed associations with eGFR. Replication was pursued in three independent multi-ancestry cohorts with and without HIV. RESULTS DNA methylation levels at 14 regions were associated with CKD. The strongest signals were located in SCARB1, DNAJC5B and C4orf50. Seven of the 14 signals also associated with eGFR, and most showed evidence for a genetic basis. Four signals (in SCARB1, FRMD4A, CSRNP1 and RAB38) replicated in other cohorts, and 11 previously reported epigenetic signals for kidney function or CKD replicated in our cohort. We found no significant DNA methylation signals in, or near, the APOL1 promoter region. CONCLUSIONS We report several novel as well as previously reported epigenetic associations with CKD and eGFR in individuals with HIV having APOL1 high-risk genotypes. Further investigation of pathways linking DNA methylation to APOL1 nephropathies is warranted.
Collapse
Affiliation(s)
- Rachel K Y Hung
- Department of HIV and Sexual Health, King's College Hospital, London, UK
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Ricardo Costeira
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Franziska Grundner-Culemann
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - John W Booth
- Department of Renal Medicine, Bart's Health NHS Foundation Trust, London, UK
| | - Claire C Sharpe
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Kate Bramham
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - Vincent C Marconi
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- School of Medicine, Emory University, Atlanta, GA, USA
| | - Alexander Teumer
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Cheryl A Winkler
- Basic Reseach Program, Frederick National Laboratory for Cancer Research and the Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Frank A Post
- Department of HIV and Sexual Health, King's College Hospital, London, UK
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Jordana T Bell
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
3
|
Di Carlo S, Longhitano E, Spinella C, Maressa V, Casuscelli C, Peritore L, Santoro D. Traditional, alternative, and emerging therapeutics for focal segmental glomerulosclerosis. Expert Opin Pharmacother 2025; 26:179-186. [PMID: 39743782 DOI: 10.1080/14656566.2024.2446621] [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: 09/11/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION Segmental focal glomerulosclerosis is a histological lesion characterized by podocyte damage. It may be a primary disease linked to an unknown circulating factor, secondary to viral infections, drug toxicity, or a disadaptive response to the loss of nephrons, or it may depend on gene mutations or have an indeterminate cause. The treatment of the primary form involves immunosuppressors. Additional pharmacotherapies for residual proteinuria are used, and emerging therapies are being studied to target other pathological pathways. AREAS COVERED This paper covers the treatment of FSGS, focusing on traditional and emerging therapeutic strategies. It is based on the KDIGO 2021 guidelines and supplemented by a literature search conducted on PubMed. EXPERT OPINION Treating FSGS is challenging due to its heterogeneity. Immunosuppression is adequate for primary FSGS but harmful in genetic or secondary forms. Key strategies include targeting the underlying cause and using agents that affect renal hemodynamics. Antifibrotic drugs can help slow kidney damage by addressing chronic inflammation and fibrosis. Alongside pharmacological treatments, managing blood pressure and restricting dietary salt are crucial. Finally, personalized treatment requires stratifying patients based on clinical, genetic, and histological data to improve clinical trial design and outcomes.
Collapse
Affiliation(s)
- Silvia Di Carlo
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. "G.Martino", University of Messina, Messina, Italy
| | - Elisa Longhitano
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. "G.Martino", University of Messina, Messina, Italy
| | - Claudia Spinella
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. "G.Martino", University of Messina, Messina, Italy
| | - Veronica Maressa
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. "G.Martino", University of Messina, Messina, Italy
| | - Chiara Casuscelli
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. "G.Martino", University of Messina, Messina, Italy
| | - Luigi Peritore
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. "G.Martino", University of Messina, Messina, Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. "G.Martino", University of Messina, Messina, Italy
| |
Collapse
|
4
|
Yu SMW, Campbell K. Genetics in Kidney Diseases. ADVANCES IN KIDNEY DISEASE AND HEALTH 2025; 32:33-40. [PMID: 40175028 PMCID: PMC11970339 DOI: 10.1053/j.akdh.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/28/2024] [Accepted: 10/08/2024] [Indexed: 04/04/2025]
Abstract
With the increasing availability of genetic testing, nephrologists can now incorporate genetic testing results into traditional biochemical profiles and kidney biopsy data to provide more accurate diagnoses. Moreover, if available, therapeutic approaches can be further tailored to individual genetic testing results to avoid unnecessary exposure to prolonged immunosuppressive medications with often undesirable side effects. Identifying new genetic mutations in kidney diseases also opens new avenues to novel drug targets. Here, we present five clinical questions summarizing some genetic kidney diseases and updates.
Collapse
Affiliation(s)
- Samuel Mon-Wei Yu
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Kirk Campbell
- Nephrology Division, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
5
|
Ray PE, Li J, Das J, Xu L, Yu J, Han Z. Pathogenesis of HIV-associated nephropathy in children and adolescents: taking a hard look 40 years later in the era of gene-environment interactions. Am J Physiol Renal Physiol 2024; 327:F1049-F1066. [PMID: 39323389 PMCID: PMC11687833 DOI: 10.1152/ajprenal.00208.2024] [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: 07/22/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024] Open
Abstract
HIV-associated nephropathy (HIVAN) is a kidney disease that affects mainly people of African ancestry with a high HIV-1 viral load. New antiretroviral therapies (ART) have been highly efficient in preventing and improving the outcome of HIVAN. However, providing chronic ART to children and adolescents living with HIV (CALWH) remains a significant challenge all over the world. More than 2.5 million CALWH, including those living in Sub-Saharan Africa, continue to be at high risk of developing HIVAN. Much of our understanding of the pathogenesis of HIVAN is based on studies conducted in transgenic mice and adults with HIVAN. However, CALWH may experience different health outcomes, risk factors, and susceptibilities to HIVAN in comparison to adults. This article reviews the progress made over the last 40 years in understanding the pathogenesis of HIVAN in CALWH, focusing on how the HIV virus, alongside genetic and environmental factors, contributes to the development of this disease. The landmark discovery that two risk alleles of the apolipoprotein-1 (APOL1) gene play a critical role in HIVAN has significantly advanced our understanding of the disease's pathogenesis. However, we still need to understand why renal inflammation persists despite ART and determine whether the kidney may harbor HIV reservoirs that need to be eliminated to cure HIV permanently. For these reasons, we emphasize reviewing how HIV-1 infects renal cells, affects their growth and regeneration, and discussing how inflammatory cytokines and APOL1 affect the outcome of childhood HIVAN.
Collapse
Affiliation(s)
- Patricio E Ray
- Department of Pediatrics and Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Jinliang Li
- Children's National Hospital, Washington, District of Columbia, United States
| | - Jharna Das
- Children's National Hospital, Washington, District of Columbia, United States
| | - Lian Xu
- Children's National Hospital, Washington, District of Columbia, United States
| | - Jing Yu
- Department of Pediatrics and Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
6
|
Gonzalez-Vicente A, Crawford DC, Bush WS, Wu Z, Bruggeman LA, Nair V, Eichinger F, Wessely O, Kretzler M, O'Toole JF, Sedor JR. Analysis of Glomerular Transcriptomes from Nephrotic Patients Suggest APOL1 Risk Variants Impact Parietal Epithelial Cells. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.05.24316766. [PMID: 39830251 PMCID: PMC11741451 DOI: 10.1101/2024.11.05.24316766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The disproportionate risk for idiopathic proteinuric podocytopathies in Black people is explained, in part, by the presence of two risk alleles (G1 or G2) in the APOL1 gene. The pathogenic mechanisms responsible for this genetic association remain incompletely understood. We analyzed glomerular RNASeq transcriptomes from patients with idiopathic nephrotic syndrome of which 72 had inferred African ancestry (AA) and 152 did not (noAA). Using gene coexpression networks we found a significant association between APOL1 risk allele number and the coexpression metamodule 2 (MM2), even after adjustment for eGFR and proteinuria at biopsy. Unadjusted Kaplan-Meier curves showed that unlike noAA, AA with the highest tertile of MM2 gene activation scores were less likely to achieve complete remission (p≤0.014). Characteristic direction (ChDir) identified a signature of 1481 genes, which separated patients with APOL1 risk alleles from those homozygous for reference APOL1 . Only in AA, the tertile with the highest activation scores of these 1481 genes was less likely to achieve complete remission (p≤0.022) and showed a trend to faster progression to the composite event of kidney failure or loss of 40% eGFR (p≤0.099). The MM2 and ChDir genes significantly overlapped and were both enriched for Epithelial Mesenchymal Transition and inflammation terms. Finally, MM2 significantly overlapped with a parietal epithelial cell (PEC)-identity gene signature but not with a podocyte identity signature. Podocytes expressing variant APOL1s may generate inflammatory signals that activate PECs by paracrine mechanisms contributing to APOL1 nephropathy.
Collapse
|
7
|
Xie Y, Liu F. Precision medicine for focal segmental glomerulosclerosis. Kidney Res Clin Pract 2024; 43:709-723. [PMID: 38325863 PMCID: PMC11615440 DOI: 10.23876/j.krcp.23.227] [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: 08/24/2023] [Revised: 11/06/2023] [Accepted: 11/15/2023] [Indexed: 02/09/2024] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is one of the common causes of nephrotic syndrome in adults and children worldwide. FSGS consists of a group of kidney diseases classified based on specific histopathological features. The current classification of FSGS makes it difficult to distinguish individual differences in pathogenesis, disease progression, and response to treatment. In recent years, the spread of next-generation sequencing, updates in biological techniques, and improvements of treatment have changed our understanding of FSGS. In this review, we will discuss the use of genetic testing in patients with FSGS, explore its clinical significance from a genetic identification perspective, and introduce several new biomarkers, that may help in the early diagnosis of FSGS and guide the development of specific or targeted therapies, so as to understand the biological characteristics in FSGS. This will certainly help develop more effective and safer treatments and advance precision medicine.
Collapse
Affiliation(s)
- Yi Xie
- Department of Nephrology, Children’s Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Liu
- Department of Nephrology, Children’s Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Yu P, Jin X, Huang W, Wang J, Zhang S, Ren L, Zhang H, Shi S. Characterization of immortalized human podocytes infected with lentivirus as an in vitro model of viral infection-associated podocytopathy. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:204-214. [PMID: 39583339 PMCID: PMC11578807 DOI: 10.62347/bbcx1142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 11/26/2024]
Abstract
A large number of studies have shown the association of kidney disease with viral infections in the body. Viral infections cause kidney injury in two manners, the systemic inflammation (cytokine storm) and the direct infection of kidney cells. Concerning direct viral infection of podocytes, the mechanism underlying virus-induced podocyte injury remains largely unknown and requires effective experimental models to facilitate its study. Here, we performed molecular characterization of immortalized human podocyte cell line (HPC) infected with lentivirus by RNA-seq. Bioinformatics analysis revealed a strong innate immune response in the cells, including interferon production and signaling. Meanwhile, activations of ferroptosis pathway and TNF-alpha signaling were also found, consistent with an impaired viability of the cells. Lentiviral infection also upregulated expression of APOL1 as observed in patients with HIV associated nephropathy (HIVAN) and diabetic nephropathy (DN). Interestingly, when the lentiviral infected cells were treated with Adriamycin (ADR), the ADR-associated signaling pathways were not interfered and remained activated as that in the cells treated with ADR only, suggesting that the virus and ADR have distinct mechanisms in damaging podocytes. Thus, the lentivirus-infected HPC cells represent a useful in vitro model of viral infection-associated podocytopathy.
Collapse
Affiliation(s)
- Peng Yu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Xi Jin
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Weijun Huang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Jingjing Wang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Sipang Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Lu Ren
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Haitao Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| | - Shaolin Shi
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Medical School of Nanjing University Nanjing 210002, Jiangsu, China
| |
Collapse
|
9
|
Meliambro K, He JC, Campbell KN. Podocyte-targeted therapies - progress and future directions. Nat Rev Nephrol 2024; 20:643-658. [PMID: 38724717 DOI: 10.1038/s41581-024-00843-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 09/14/2024]
Abstract
Podocytes are the key target cells for injury across the spectrum of primary and secondary proteinuric kidney disorders, which account for up to 90% of cases of kidney failure worldwide. Seminal experimental and clinical studies have established a causative link between podocyte depletion and the magnitude of proteinuria in progressive glomerular disease. However, no substantial advances have been made in glomerular disease therapies, and the standard of care for podocytopathies relies on repurposed immunosuppressive drugs. The past two decades have seen a remarkable expansion in understanding of the mechanistic basis of podocyte injury, with prospects increasing for precision-based treatment approaches. Dozens of disease-causing genes with roles in the pathogenesis of clinical podocytopathies have been identified, as well as a number of putative glomerular permeability factors. These achievements, together with the identification of novel targets of podocyte injury, the development of potential approaches to harness the endogenous podocyte regenerative potential of progenitor cell populations, ongoing clinical trials of podocyte-specific pharmacological agents and the development of podocyte-directed drug delivery systems, contribute to an optimistic outlook for the future of glomerular disease therapy.
Collapse
Affiliation(s)
- Kristin Meliambro
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirk N Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Olabisi OA, Barrett NJ, Lucas A, Smith M, Bethea K, Soldano K, Croall S, Sadeghpour A, Chakraborty H, Wolf M. Design and Rationale of the Phase 2 Baricitinib Study in Apolipoprotein L1-Mediated Kidney Disease (JUSTICE). Kidney Int Rep 2024; 9:2677-2684. [PMID: 39291185 PMCID: PMC11403079 DOI: 10.1016/j.ekir.2024.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Individuals of recent West African ancestry develop focal segmental glomerulosclerosis (FSGS) and hypertension-attributed end-stage kidney disease (HTN-ESKD) at 4 times the rate of White Americans. Two protein-coding variants of the Apolipoprotein L1 (APOL1) gene, G1 and G2, explain 50% to 70% of the excess risk of HTN-ESKD and FSGS among this group. Increased expression of G1 and G2 in the kidney, mediated by Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling, drive pathogenesis of these kidney diseases. Baricitinib is an orally active inhibitor of JAK1/2 that blocks APOL1 synthesis. The Janus kinase-STAT Inhibition to Reduce APOL1-Associated Kidney Disease (JUSTICE) trial is evaluating the antiproteinuric efficacy and safety of baricitinib in patients with APOL1-associated FSGS and HTN-attributed chronic kidney disease (HTN-CKD). Methods JUSTICE is a single-center, randomized, double-blind, placebo-controlled, pilot phase 2 trial of baricitinib in patients with proteinuria, APOL1-associated FSGS or APOL1-associated HTN-CKD without diabetes. A total of 75 African American patients with APOL1-associated CKD, including 25 with FSGS and 50 with HTN-CKD, aged 18 to 70 years will be randomized 2:1 to daily treatment with baricitinib or placebo, respectively. Results The primary efficacy end point will be percent change in urine albumin-to-creatinine ratio (UACR) from baseline to end of month 6. The primary safety end point will be incidence of clinically significant decreases in hemoglobin of ≥ 1g/dl. Conclusion The phase 2 JUSTICE study will characterize the antiproteinuric efficacy and safety of JAK1/2 inhibition with baricitinib in patients with APOL1-associated FSGS and APOL1-associated HTN-CKD.
Collapse
Affiliation(s)
- Opeyemi A Olabisi
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Nadine J Barrett
- Atrium Health/Wake Forest Comprehensive Cancer Center and Maya Angelo Center for Health Equity, Wake Forest School of Medicine, Wake Forest, North Carolina, USA
- Department of Social Science and Health Policy, Division of Population Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Maya Angelo Center for Health Equity, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anika Lucas
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Maurice Smith
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kenisha Bethea
- Duke Clinical and Translational Science Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Karen Soldano
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Stephanie Croall
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Azita Sadeghpour
- Duke Precision Medicine Program, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Myles Wolf
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
11
|
Nystrom SE, Soldano KL, Rockett M, Datta S, Li G, Silas D, Garrett ME, Ashley-Koch AE, Olabisi OA. APOL1 High-Risk Genotype is Not Associated With New or Worsening of Proteinuria or Kidney Function Decline Following COVID-19 Vaccination. Kidney Int Rep 2024; 9:2657-2666. [PMID: 39291186 PMCID: PMC11403097 DOI: 10.1016/j.ekir.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction SARS-CoV-2 infection increases systemic inflammatory cytokines which act as a second-hit driver of Apolipoprotein L1 (APOL1)-mediated collapsing glomerulopathy. SARS-CoV-2 vaccination also increases cytokines. Recent reports of new glomerular disease in individuals with APOL1 high-risk genotype (HRG) following SARS-CoV-2 vaccination raised the concern SARS-CoV-2 vaccination may also act as a second-hit driver of APOL1-mediated glomerulopathy. Methods We screened 1507 adults in the Duke's Measurement to Understand Reclassification of Disease of Cabarrus and Kannapolis (MURDOCK) registry and enrolled 105 eligible participants with available SARS-CoV-2 vaccination data, prevaccination and postvaccination serum creatinine, and urine protein measurements. Paired data were stratified by number of APOL1 risk alleles (RAs) and compared within groups using Wilcoxon signed rank test and across groups by analysis of variance. Results Among 105 participants, 30 (28.6%) had 2, 39 (37.1%) had 1, and 36 (34.3%) had 0 APOL1 RA. Most of the participants (94%) received at least 2 doses of vaccine. Most (98%) received the BNT162B2 (Pfizer) or mRNA-1273 (Moderna) vaccine. On average, the prevaccine and postvaccine laboratory samples were drawn 648 days apart. There were no detectable differences between pre- and post-serum creatinine or pre- and post-urine albumin creatinine ratio irrespective of the participants' APOL1 genotype. Finally, most participants with APOL1 RA had the most common haplotype (E150, I228, and K255) and lacked the recently described protective N264K haplotype. Conclusion In this observational study, APOL1 HRG is not associated with new or worsening of proteinuria or decline in kidney function following SARS-CoV-2 vaccination. Validation of this result in larger cohorts would further support the renal safety of SARS-CoV-2 vaccine in individuals with APOL1 HRG.
Collapse
Affiliation(s)
- Sarah E Nystrom
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Karen L Soldano
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Micki Rockett
- Duke Clinical and Translational Science Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Somenath Datta
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Guojie Li
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Daniel Silas
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Melanie E Garrett
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Allison E Ashley-Koch
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Opeyemi A Olabisi
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Suresh V, Stillman IE, Campbell KN, Meliambro K. Focal Segmental Glomerulosclerosis. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:275-289. [PMID: 39084753 DOI: 10.1053/j.akdh.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 08/02/2024]
Abstract
Focal segmental glomerular sclerosis (FSGS) is a histological lesion characterized by sclerosis in sections (segmental) of some glomeruli (focal) in association with podocyte injury. Historically, FSGS has often been characterized as a disease, but it is a heterogeneous entity based on etiology, clinical course, and therapeutic approach. A unifying feature is podocyte injury and loss, which can be primary or the result of secondary maladaptive responses to glomerular stressors. FSGS has been demonstrated over time to carry a large health burden and remains a leading glomerular cause of ESRD globally. Recent clinical practice guidelines highlight the unmet scientific need for better understanding of disease pathogenesis, particularly for immunologic etiologies, as well as more targeted therapeutic drug development. In this review, we will discuss the current FSGS classification scheme, pathophysiologic mechanisms of injury, and treatment guidelines, along with emerging and investigational therapeutics.
Collapse
Affiliation(s)
- Varsha Suresh
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Isaac E Stillman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kirk N Campbell
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Kristin Meliambro
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
15
|
Juliar BA, Stanaway IB, Sano F, Fu H, Smith KD, Akilesh S, Scales SJ, El Saghir J, Bhatraju PK, Liu E, Yang J, Lin J, Eddy S, Kretzler M, Zheng Y, Himmelfarb J, Harder JL, Freedman BS. Interferon-γ induces combined pyroptotic angiopathy and APOL1 expression in human kidney disease. Cell Rep 2024; 43:114310. [PMID: 38838223 PMCID: PMC11216883 DOI: 10.1016/j.celrep.2024.114310] [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: 09/27/2023] [Revised: 03/18/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024] Open
Abstract
Elevated interferon (IFN) signaling is associated with kidney diseases including COVID-19, HIV, and apolipoprotein-L1 (APOL1) nephropathy, but whether IFNs directly contribute to nephrotoxicity remains unclear. Using human kidney organoids, primary endothelial cells, and patient samples, we demonstrate that IFN-γ induces pyroptotic angiopathy in combination with APOL1 expression. Single-cell RNA sequencing, immunoblotting, and quantitative fluorescence-based assays reveal that IFN-γ-mediated expression of APOL1 is accompanied by pyroptotic endothelial network degradation in organoids. Pharmacological blockade of IFN-γ signaling inhibits APOL1 expression, prevents upregulation of pyroptosis-associated genes, and rescues vascular networks. Multiomic analyses in patients with COVID-19, proteinuric kidney disease, and collapsing glomerulopathy similarly demonstrate increased IFN signaling and pyroptosis-associated gene expression correlating with accelerated renal disease progression. Our results reveal that IFN-γ signaling simultaneously induces endothelial injury and primes renal cells for pyroptosis, suggesting a combinatorial mechanism for APOL1-mediated collapsing glomerulopathy, which can be targeted therapeutically.
Collapse
Affiliation(s)
- Benjamin A Juliar
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Ian B Stanaway
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Fumika Sano
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Hongxia Fu
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Division of Hematology, Department of Medicine, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA; Bloodworks Northwest Research Institute, Seattle, WA 98102, USA; Plurexa, Seattle, WA 98109, USA
| | - Kelly D Smith
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Shreeram Akilesh
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Suzie J Scales
- Department of Immunology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jamal El Saghir
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pavan K Bhatraju
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Esther Liu
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Johnson Yang
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jennie Lin
- Division of Nephrology and Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sean Eddy
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ying Zheng
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jennifer L Harder
- Division of Nephrology, Department of Internal Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Benjamin S Freedman
- Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA; Plurexa, Seattle, WA 98109, USA.
| |
Collapse
|
16
|
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
|
17
|
Koehler S, Hengel FE, Dumoulin B, Damashek L, Holzman LB, Susztak K, Huber TB. The 14th International Podocyte Conference 2023: from podocyte biology to glomerular medicine. Kidney Int 2024; 105:935-952. [PMID: 38447880 DOI: 10.1016/j.kint.2024.01.042] [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: 10/30/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 03/08/2024]
Abstract
The 14th International Podocyte Conference took place in Philadelphia, Pennsylvania, USA from May 23 to 26, 2023. It commenced with an early-career researchers' meeting on May 23, providing young scientists with a platform to present and discuss their research findings. Throughout the main conference, 29 speakers across 9 sessions shared their insights on podocyte biology, glomerular medicine, novel technologic advancements, and translational approaches. Additionally, the event featured 3 keynote lectures addressing engineered chimeric antigen receptor T cell- and mRNA-based therapies and the use of biobanks for enhanced disease comprehension. Furthermore, 4 brief oral abstract sessions allowed scientists to present their findings to a broad audience. The program also included a panel discussion addressing the challenges of conducting human research within the American Black community. Remarkably, after a 5-year hiatus from in-person conferences, the 14th International Podocyte Conference successfully convened scientists from around the globe, fostering the presentation and discussion of crucial research findings, as summarized in this review. Furthermore, to ensure continuous and sustainable education, research, translation, and trial medicine related to podocyte and glomerular diseases for the benefit of patients, the International Society of Glomerular Disease was officially launched during the conference.
Collapse
Affiliation(s)
- Sybille Koehler
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Felicitas E Hengel
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Bernhard Dumoulin
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Laurel Damashek
- International Society of Glomerular Disease, Florence, Massachusetts, USA
| | - Lawrence B Holzman
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tobias B Huber
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; International Society of Glomerular Disease, Florence, Massachusetts, USA.
| |
Collapse
|
18
|
Park SG, Keller A, Kaiser NK, Bruce JE. Interactome dynamics during heat stress signal transmission and reception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591712. [PMID: 38746244 PMCID: PMC11092488 DOI: 10.1101/2024.04.29.591712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Among evolved molecular mechanisms, cellular stress response to altered environmental conditions to promote survival is among the most fundamental. The presence of stress-induced unfolded or misfolded proteins and molecular registration of these events constitute early steps in cellular stress response. However, what stress-induced changes in protein conformations and protein-protein interactions within cells initiate stress response and how these features are recognized by cellular systems are questions that have remained difficult to answer, requiring new approaches. Quantitative in vivo chemical cross-linking coupled with mass spectrometry (qXL-MS) is an emerging technology that provides new insight on protein conformations, protein-protein interactions and how the interactome changes during perturbation within cells, organelles, and even tissues. In this work, qXL-MS and quantitative proteome analyses were applied to identify significant time-dependent interactome changes that occur prior to large-scale proteome abundance remodeling within cells subjected to heat stress. Interactome changes were identified within minutes of applied heat stress, including stress-induced changes in chaperone systems as expected due to altered functional demand. However, global analysis of all interactome changes revealed the largest significant enrichment in the gene ontology molecular function term of RNA binding. This group included more than 100 proteins among multiple components of protein synthesis machinery, including mRNA binding, spliceosomes, and ribosomes. These interactome data provide new conformational insight on the complex relationship that exists between transcription, translation and cellular stress response mechanisms. Moreover, stress-dependent interactome changes suggest that in addition to conformational stabilization of RNA-binding proteins, adaptation of RNA as interacting ligands offers an additional fitness benefit resultant from generally lower RNA thermal stability. As such, RNA ligands also serve as fundamental temperature sensors that signal stress through decreased conformational regulation of their protein partners as was observed in these interactome dynamics.
Collapse
|
19
|
Kovalik ME, Dacanay MA, Crowley SD, Hall G. Swollen Feet: Considering the Paradoxical Roles of Interleukins in Nephrotic Syndrome. Biomedicines 2024; 12:738. [PMID: 38672094 PMCID: PMC11048099 DOI: 10.3390/biomedicines12040738] [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: 01/26/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/28/2024] Open
Abstract
Interleukins are a family of 40 bioactive peptides that act through cell surface receptors to induce a variety of intracellular responses. While interleukins are most commonly associated with destructive, pro-inflammatory signaling in cells, some also play a role in promoting cellular resilience and survival. This review will highlight recent evidence of the cytoprotective actions of the interleukin 1 receptor (IL-1R)- and common gamma chain receptor (IL-Rγc)-signaling cytokines in nephrotic syndrome (NS). NS results from the injury or loss of glomerular visceral epithelial cells (i.e., podocytes). Although the causes of podocyte dysfunction vary, it is clear that pro-inflammatory cytokines play a significant role in regulating the propagation, duration and severity of disease. Pro-inflammatory cytokines signaling through IL-1R and IL-Rγc have been shown to exert anti-apoptotic effects in podocytes through the phosphoinositol-3-kinase (PI-3K)/AKT pathway, highlighting the potential utility of IL-1R- and IL-Rγc-signaling interleukins for the treatment of podocytopathy in NS. The paradoxical role of interleukins as drivers and mitigators of podocyte injury is complex and ill-defined. Emerging evidence of the cytoprotective role of some interleukins in NS highlights the urgent need for a nuanced understanding of their pro-survival benefits and reveals their potential as podocyte-sparing therapeutics for NS.
Collapse
Affiliation(s)
- Maria E. Kovalik
- Division of Nephrology, Duke University, Durham, NC 27701, USA; (M.E.K.)
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Monique A. Dacanay
- Division of Nephrology, Duke University, Durham, NC 27701, USA; (M.E.K.)
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Steven D. Crowley
- Division of Nephrology, Duke University, Durham, NC 27701, USA; (M.E.K.)
| | - Gentzon Hall
- Division of Nephrology, Duke University, Durham, NC 27701, USA; (M.E.K.)
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| |
Collapse
|
20
|
Anandakrishnan N, Yi Z, Sun Z, Liu T, Haydak J, Eddy S, Jayaraman P, DeFronzo S, Saha A, Sun Q, Yang D, Mendoza A, Mosoyan G, Wen HH, Schaub JA, Fu J, Kehrer T, Menon R, Otto EA, Godfrey B, Suarez-Farinas M, Leffters S, Twumasi A, Meliambro K, Charney AW, García-Sastre A, Campbell KN, Gusella GL, He JC, Miorin L, Nadkarni GN, Wisnivesky J, Li H, Kretzler M, Coca SG, Chan L, Zhang W, Azeloglu EU. Integrated multiomics implicates dysregulation of ECM and cell adhesion pathways as drivers of severe COVID-associated kidney injury. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.18.24304401. [PMID: 38562892 PMCID: PMC10984064 DOI: 10.1101/2024.03.18.24304401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
COVID-19 has been a significant public health concern for the last four years; however, little is known about the mechanisms that lead to severe COVID-associated kidney injury. In this multicenter study, we combined quantitative deep urinary proteomics and machine learning to predict severe acute outcomes in hospitalized COVID-19 patients. Using a 10-fold cross-validated random forest algorithm, we identified a set of urinary proteins that demonstrated predictive power for both discovery and validation set with 87% and 79% accuracy, respectively. These predictive urinary biomarkers were recapitulated in non-COVID acute kidney injury revealing overlapping injury mechanisms. We further combined orthogonal multiomics datasets to understand the mechanisms that drive severe COVID-associated kidney injury. Functional overlap and network analysis of urinary proteomics, plasma proteomics and urine sediment single-cell RNA sequencing showed that extracellular matrix and autophagy-associated pathways were uniquely impacted in severe COVID-19. Differentially abundant proteins associated with these pathways exhibited high expression in cells in the juxtamedullary nephron, endothelial cells, and podocytes, indicating that these kidney cell types could be potential targets. Further, single-cell transcriptomic analysis of kidney organoids infected with SARS-CoV-2 revealed dysregulation of extracellular matrix organization in multiple nephron segments, recapitulating the clinically observed fibrotic response across multiomics datasets. Ligand-receptor interaction analysis of the podocyte and tubule organoid clusters showed significant reduction and loss of interaction between integrins and basement membrane receptors in the infected kidney organoids. Collectively, these data suggest that extracellular matrix degradation and adhesion-associated mechanisms could be a main driver of COVID-associated kidney injury and severe outcomes.
Collapse
|
21
|
Ma K, Luo L, Yang M, Meng Y. The suppression of sepsis-induced kidney injury via the knockout of T lymphocytes. Heliyon 2024; 10:e23311. [PMID: 38283245 PMCID: PMC10818183 DOI: 10.1016/j.heliyon.2023.e23311] [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: 05/09/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 01/30/2024] Open
Abstract
Patients with sepsis always have a high mortality rate, and acute kidney injury (AKI) is the main cause of death. It seems obvious that the immune response is involved in this process, but the specific mechanism is unknown, especially the pathogenic role of T cells and B cells needs to be further clarified. Acute kidney injury models induced by lipopolysaccharide were established using T-cell, B-cell, and T&B cell knockout mice to elucidate the role of immune cells in sepsis. Flow cytometry was used to validate the mouse models, and the pathology can confirm renal tubular injury. LPS-induced sepsis caused significant renal pathological damage, Second-generation gene sequencing showed T cells-associated pathway was enriched in sepsis. The renal tubular injury was significantly reduced in T cell and T&B cell knockout mice (BALB/c-nu, Rag1-/-), especially in BALB/c-nu mice, with a decrease in the secretion of inflammatory cytokines in the renal tissue after LPS injection. LPS injection did not produce the same effect after the knockout of B cells. We found that blocking T cells could alleviate inflammation and renal injury caused by sepsis, providing a promising strategy for controlling renal injury.
Collapse
Affiliation(s)
- Ke Ma
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
| | - Liang Luo
- The Biomedical Translational Research Institute, Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University), Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Meixiang Yang
- The Biomedical Translational Research Institute, Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University), Guangzhou Key Laboratory for Germ-free Animals and Microbiota Application, School of Medicine, Jinan University, Guangzhou, 510632, China
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Yu Meng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
- Department of Nephrology, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Heyuan, 517000, China
| |
Collapse
|
22
|
Chau CW, Sugimura R. Organoids in COVID-19: can we break the glass ceiling? J Leukoc Biol 2024; 115:85-99. [PMID: 37616269 DOI: 10.1093/jleuko/qiad098] [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: 01/30/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
COVID-19 emerged in September 2020 as a disease caused by the virus SARS-CoV-2. The disease presented as pneumonia at first but later was shown to cause multisystem infections and long-term complications. Many efforts have been put into discovering the exact pathogenesis of the disease. In this review, we aim to discuss an emerging tool in disease modeling, organoids, in the investigation of COVID-19. This review will introduce some methods and breakthroughs achieved by organoids and the limitations of this system.
Collapse
Affiliation(s)
- Chiu Wang Chau
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, 21 Sassoon Rd, Pokfulam 99077, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, 21 Sassoon Rd, Pokfulam 99077, Hong Kong
- Centre for Translational Stem Cell Biology, 17 Science Park W Ave, Science Park 999077, Hong Kong
| |
Collapse
|
23
|
Sedor JR. APOL1 Kidney Disease: Discovery to Targeted Therapy in 10 Years. Clin J Am Soc Nephrol 2024; 19:126-128. [PMID: 37459117 PMCID: PMC10843197 DOI: 10.2215/cjn.0000000000000256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Affiliation(s)
- John R Sedor
- Glickman Urology and Kidney and Lerner Research Institutes , Cleveland Clinic , and Departments of Molecular Medicine and Physiology and Biophysics , Case Western Reserve University, Cleveland , Ohio
| |
Collapse
|
24
|
Vasquez-Rios G, De Cos M, Campbell KN. Novel Therapies in APOL1-Mediated Kidney Disease: From Molecular Pathways to Therapeutic Options. Kidney Int Rep 2023; 8:2226-2234. [PMID: 38025220 PMCID: PMC10658239 DOI: 10.1016/j.ekir.2023.08.028] [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: 08/05/2023] [Accepted: 08/21/2023] [Indexed: 12/01/2023] Open
Abstract
Apolipoprotein L1 (APOL1) high-risk variants confer an increased risk for the development and progression of kidney disease among individuals of recent African ancestry. Over the past several years, significant progress has been made in understanding the pathogenesis of APOL1-mediated kidney diseases (AMKD), including genetic regulation, environmental interactions, immunomodulatory, proinflammatory and apoptotic signaling processes, as well as the complex role of APOL1 as an ion channel. Collectively, these findings have paved the way for novel therapeutic strategies to mitigate APOL1-mediated kidney injury. Precision medicine approaches are being developed to identify subgroups of AMKD patients who may benefit from these targeted interventions, fueling hope for improved clinical outcomes. This review summarizes key mechanistic insights in the pathogenesis of AMKD, emergent therapies, and discusses future challenges.
Collapse
Affiliation(s)
- George Vasquez-Rios
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marina De Cos
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirk N. Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
25
|
An Y, He Y, Ge N, Guo J, Yang F, Sun S. Organoids to Remodel SARS-CoV-2 Research: Updates, Limitations and Perspectives. Aging Dis 2023; 14:1677-1699. [PMID: 37196111 PMCID: PMC10529756 DOI: 10.14336/ad.2023.0209] [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: 01/11/2023] [Accepted: 02/09/2023] [Indexed: 05/19/2023] Open
Abstract
The novel COVID-19 pneumonia caused by the SARS-CoV-2 virus poses a significant threat to human health. Scientists have made significant efforts to control this virus, consequently leading to the development of novel research methods. Traditional animal and 2D cell line models might not be suitable for large-scale applications in SARS-CoV-2 research owing to their limitations. As an emerging modelling method, organoids have been applied in the study of various diseases. Their advantages include their ability to closely mirror human physiology, ease of cultivation, low cost, and high reliability; thus, they are considered to be a suitable choice to further the research on SARS-CoV-2. During the course of various studies, SARS-CoV-2 was shown to infect a variety of organoid models, exhibiting changes similar to those observed in humans. This review summarises the various organoid models used in SARS-CoV-2 research, revealing the molecular mechanisms of viral infection and exploring the drug screening tests and vaccine research that have relied on organoid models, hence illustrating the role of organoids in remodelling SARS-CoV-2 research.
Collapse
Affiliation(s)
- Yucheng An
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Yanjie He
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York, NY, USA
| | - Nan Ge
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Jintao Guo
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Fan Yang
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| | - Siyu Sun
- Department of Gastroenterology, Shengjing hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Casuscelli C, Longhitano E, Maressa V, Di Carlo S, Peritore L, Di Lorenzo S, Calabrese V, Cernaro V, Santoro D. Autoimmunity and Infection in Glomerular Disease. Microorganisms 2023; 11:2227. [PMID: 37764071 PMCID: PMC10538233 DOI: 10.3390/microorganisms11092227] [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: 07/26/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The ongoing glomerular damage of infections is not limited to the most widely known form of post-streptococcal glomerulonephritis, which is today less common in the Western world; other forms of glomerulonephritis are associated with several bacterial, viral and parasitic pathogens. The mechanisms responsible range from the direct damage of glomerular cells to the formation and deposition of immunocomplexes to molecular mimicry to the secretion of superantigens. Similarly, in the course of glomerular disease, infections are more frequent than in the general population due to the loss of immunoglobulins in urine and the immunosuppressive agents used to treat the autoimmune disease that decrease the activity of the immune system. Recognizing this two-way link, understanding its pathogenetic mechanism, and identifying the most appropriate therapeutic choice are essential for the personalized management of patients. In this continuously developing field, this short review summarizes the current state of the art as support for physicians, who are increasingly involved in managing patients with glomerular disease and infections.
Collapse
Affiliation(s)
- Chiara Casuscelli
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98122 Messina, Italy; (E.L.); (V.M.); (S.D.C.); (L.P.); (S.D.L.); (V.C.); (V.C.)
| | | | | | | | | | | | | | | | - Domenico Santoro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U. “G. Martino”, University of Messina, 98122 Messina, Italy; (E.L.); (V.M.); (S.D.C.); (L.P.); (S.D.L.); (V.C.); (V.C.)
| |
Collapse
|
27
|
Pell J, Nagata S, Menon MC. Nonpodocyte Roles of APOL1 Variants: An Evolving Paradigm. KIDNEY360 2023; 4:e1325-e1331. [PMID: 37461136 PMCID: PMC10550003 DOI: 10.34067/kid.0000000000000216] [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: 05/06/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
Since the seminal discovery of the trypanolytic, exonic variants in apolipoprotein L1 (APOL1) and their association with kidney disease in individuals of recent African ancestry, a wide body of research has emerged offering key insights into the mechanisms of disease. Importantly, the podocyte has become a focal point for our understanding of how risk genotype leads to disease, with activation of putative signaling pathways within the podocyte identified as playing a causal role in podocytopathy, FSGS, and progressive renal failure. However, the complete mechanism of genotype-to-phenotype progression remains incompletely understood in APOL1-risk individuals. An emerging body of evidence reports more than podocyte-intrinsic expression of APOL1 risk variants is needed for disease to manifest. This article reviews the seminal data and reports which placed the podocyte at the center of our understanding of APOL1-FSGS, as well as the evident shortcomings of this podocentric paradigm. We examine existing evidence for environmental and genetic factors that may influence disease, drawing from both clinical data and APOL1's fundamental role as an immune response gene. We also review the current body of data for APOL1's impact on nonpodocyte cells, including endothelial cells, the placenta, and immune cells in both a transplant and native setting. Finally, we discuss the implications of these emerging data and how the paradigm of disease might evolve as a result.
Collapse
Affiliation(s)
- John Pell
- Department of Medicine , Yale University, New Haven , Connecticut
| | | | | |
Collapse
|
28
|
Olabisi OA. APOL1 channel blocker reduces proteinuria in FSGS. Kidney Int 2023; 104:228-230. [PMID: 37224918 PMCID: PMC11044920 DOI: 10.1016/j.kint.2023.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/26/2023]
Affiliation(s)
- Opeyemi A Olabisi
- Department of Medicine, Division of Nephrology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
29
|
Anders HJ, Kitching AR, Leung N, Romagnani P. Glomerulonephritis: immunopathogenesis and immunotherapy. Nat Rev Immunol 2023; 23:453-471. [PMID: 36635359 PMCID: PMC9838307 DOI: 10.1038/s41577-022-00816-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 01/14/2023]
Abstract
'Glomerulonephritis' (GN) is a term used to describe a group of heterogeneous immune-mediated disorders characterized by inflammation of the filtration units of the kidney (the glomeruli). These disorders are currently classified largely on the basis of histopathological lesion patterns, but these patterns do not align well with their diverse pathological mechanisms and hence do not inform optimal therapy. Instead, we propose grouping GN disorders into five categories according to their immunopathogenesis: infection-related GN, autoimmune GN, alloimmune GN, autoinflammatory GN and monoclonal gammopathy-related GN. This categorization can inform the appropriate treatment; for example, infection control for infection-related GN, suppression of adaptive immunity for autoimmune GN and alloimmune GN, inhibition of single cytokines or complement factors for autoinflammatory GN arising from inborn errors in innate immunity, and plasma cell clone-directed or B cell clone-directed therapy for monoclonal gammopathies. Here we present the immunopathogenesis of GN and immunotherapies in use and in development and discuss how an immunopathogenesis-based GN classification can focus research, and improve patient management and teaching.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University Munich, Munich, Germany.
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Department of Paediatric Nephrology, Monash Health, Clayton, VIC, Australia
| | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Paola Romagnani
- Department of Experimental and Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| |
Collapse
|
30
|
Medina E, Rueda C, Batlle D. FSGS and COVID-19 in Non-African American Patients. KIDNEY360 2023; 4:687-699. [PMID: 37229730 PMCID: PMC10371264 DOI: 10.34067/kid.0000000000000104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/10/2023] [Indexed: 05/27/2023]
Abstract
Collapsing Focal Segmental Glomerulosclerosis (FSGS) has been reported relatively frequently in African American (AA) patients with coronavirus disease 2019 (COVID-19), and it is associated almost always with Apolipoprotein L gen 1 (APOL1) high-risk variants. We reviewed the published literature from April 2020 to November 2022 searching for non-African American (non-AA) patients with FSGS associated with COVID-19 (eight White patients, six Hispanic patients, three Asian patients, one Indian patient, and one Asian Indian patient). The following histologic patterns were found: collapsing (n=11), not otherwise specified (n=5), tip (n=2), and perihilar (n=1). Fifteen of the 19 patients had AKI. The APOL1 genotype was reported in only six of the 19 non-AA patients. Three of them (two Hispanic patients and one White patient) with collapsing FSGS had high-risk APOL1 variants. The other three patients (two White patients and one Hispanic patient with the collapsing variant, tip variant, and not otherwise specified) had low-risk APOL1 variants. Among 53 African American patients with collapsing FSGS associated with COVID-19, 48 had high-risk APOL1 variants and five had low-risk APOL1 variants. We conclude that in non-AA patients, FSGS is a rare complication of COVID-19. FSGS associated with COVID-19 can occur rarely with low-risk APOL1 variants in non-AA and AA patients. Non-AA patients reported to be associated with high-risk APOL1 variants possibly reflect inaccuracy of self-reported race with AA admixture because of unknown ancestry. Given the importance of APOL1 in the pathogenesis of FSGS associated with viral infection and to avoid racial bias, it seems appropriate that APOL1 testing be considered in patients with FSGS associated with COVID-19, regardless of self-reported race.
Collapse
Affiliation(s)
- Elba Medina
- Division of Nephrology, General Hospital of México, Eduardo Liceaga, México City, México
- Master's and PhD Program in Dental and Health Medical Sciences, Universidad Nacional Autónoma de México, México City, México
| | - Carlos Rueda
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Daniel Batlle
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
31
|
Ferrarini A, Vacca A, Solimando AG, Tavio M, Acquaviva R, Rocchi M, Nitti C, Salvi A, Menditto V, Luchetti Gentiloni MM, Russo A, Moretti M, Pavani M, Giacometti A, Bonifazi M, Zuccatosta L, Romani L, Racanelli V, Moroncini G, Gabrielli A, Pomponio G. Early administration of tofacitinib in COVID-19 pneumonitis: An open randomised controlled trial. Eur J Clin Invest 2023; 53:e13898. [PMID: 36380693 DOI: 10.1111/eci.13898] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Controversies on sub-populations most sensitive to therapy and the best timing of starting the treatment still surround the use of immunomodulatory drugs in COVID-19. OBJECTIVES We designed a multicentre open-label randomised controlled trial to test the effect of prompt adding of tofacitinib to standard therapy for hospitalised patients affected by mild/moderate COVID-19 pneumonitis. METHODS Patients admitted to three Italian hospitals affected by COVID-19 pneumonitis not requiring mechanical ventilation were randomised to receive standard treatment alone or tofacitinib (10 mg/bid) for 2 weeks, starting within the first 24 h from admission. RESULTS A total of 116 patients were randomised; 49 in the experimental arm completed the 14-day treatment period, 9 discontinued tofacitinib as the disease worsened and were included in the analysis, and 1 died of respiratory failure. All 58 control patients completed the study. Clinical and demographic characteristics were similar between the study groups. In the tofacitinib group, 9/58 (15.5%) patients progressed to noninvasive ventilation (CPAP) to maintain SO2 > 93%, invasive mechanical ventilation or death by day 14 was 15.5%, significantly less than in the control group (20/58, 34.4%, RR 0,45, RRR -55%, NNT 5; p = .018). No differences in severe adverse effect incidence had been observed across the groups. CONCLUSION High-dose tofacitinib therapy in patients with COVID pneumonitis is safe and may prevent deterioration to respiratory failure.
Collapse
Affiliation(s)
- Alessia Ferrarini
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Gastroenterologia ed Endoscopia Digestiva, Ospedali Riuniti Marche Nord, Fano, Italy
| | - Angelo Vacca
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Antonio Giovanni Solimando
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy.,IRCCS Istituto Tumori "Giovanni Paolo II" Bari, Bari, Italy
| | - Marcello Tavio
- Malattie Infettive, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Rossella Acquaviva
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Marco Rocchi
- Statistica Medica, Dipartimento di Scienze Biomolecolari, Università di Urbino, Urbino, Italy
| | - Cinzia Nitti
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Aldo Salvi
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vincenzo Menditto
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | - Alessandro Russo
- Clinica di Malattie Infettive e Tropicali Dipartimento di Scienze Mediche e Chirurgiche Università "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Marco Moretti
- SOD Medicina di Laboratorio Ospedali Riuniti di Ancona, Ancona, Italy
| | - Marianna Pavani
- SOD Medicina di Laboratorio Ospedali Riuniti di Ancona, Ancona, Italy
| | - Andrea Giacometti
- Clinica di Malattie Infettive, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | | | - Laura Romani
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vito Racanelli
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Gianluca Moroncini
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Armando Gabrielli
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | | |
Collapse
|
32
|
Jain NK, Tailang M, Jain HK, Chandrasekaran B, Sahoo BM, Subramanian A, Thangavel N, Aldahish A, Chidambaram K, Alagusundaram M, Kumar S, Selvam P. Therapeutic implications of current Janus kinase inhibitors as anti-COVID agents: A review. Front Pharmacol 2023; 14:1135145. [PMID: 37021053 PMCID: PMC10067607 DOI: 10.3389/fphar.2023.1135145] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
Severe cases of COVID-19 are characterized by hyperinflammation induced by cytokine storm, ARDS leading to multiorgan failure and death. JAK-STAT signaling has been implicated in immunopathogenesis of COVID-19 infection under different stages such as viral entry, escaping innate immunity, replication, and subsequent inflammatory processes. Prompted by this fact and prior utilization as an immunomodulatory agent for several autoimmune, allergic, and inflammatory conditions, Jakinibs have been recognized as validated small molecules targeting the rapid release of proinflammatory cytokines, primarily IL-6, and GM-CSF. Various clinical trials are under investigation to evaluate Jakinibs as potential candidates for treating COVID-19. Till date, there is only one small molecule Jakinib known as baricitinib has received FDA-approval as a standalone immunomodulatory agent in treating critical COVID-19 patients. Though various meta-analyses have confirmed and validated the safety and efficacy of Jakinibs, further studies are required to understand the elaborated pathogenesis of COVID-19, duration of Jakinib treatment, and assess the combination therapeutic strategies. In this review, we highlighted JAK-STAT signalling in the pathogenesis of COVID-19 and clinically approved Jakinibs. Moreover, this review described substantially the promising use of Jakinibs and discussed their limitations in the context of COVID-19 therapy. Hence, this review article provides a concise, yet significant insight into the therapeutic implications of Jakinibs as potential anti-COVID agents which opens up a new horizon in the treatment of COVID-19, effectively.
Collapse
Affiliation(s)
- Nem Kumar Jain
- School of Pharmacy, ITM University, Gwalior, Madhya Pradesh, India
- School of Studies in Pharmaceutical Sciences, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Mukul Tailang
- School of Studies in Pharmaceutical Sciences, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Hemant Kumar Jain
- Department of General Medicine, Government Medical College, Datia, Madhya Pradesh, India
| | - Balakumar Chandrasekaran
- Faculty of Pharmacy, Philadelphia University, Amman, Jordan
- *Correspondence: Balakumar Chandrasekaran, ; Palani Selvam,
| | - Biswa Mohan Sahoo
- Roland Institute of Pharmaceutical Sciences, Berhampur, Odisha, India
| | - Anandhalakshmi Subramanian
- Department of Microbiology and Clinical Parasitology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Neelaveni Thangavel
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Afaf Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - M. Alagusundaram
- School of Pharmacy, ITM University, Gwalior, Madhya Pradesh, India
| | - Santosh Kumar
- School of Sciences, ITM University, Gwalior, Madhya Pradesh, India
| | - Palani Selvam
- School of Medicine, College of Medicine and Health Sciences, Jijiga University, Jijiga, Ethiopia
- *Correspondence: Balakumar Chandrasekaran, ; Palani Selvam,
| |
Collapse
|
33
|
Lei S, Chen X, Wu J, Duan X, Men K. Small molecules in the treatment of COVID-19. Signal Transduct Target Ther 2022; 7:387. [PMID: 36464706 PMCID: PMC9719906 DOI: 10.1038/s41392-022-01249-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 12/11/2022] Open
Abstract
The outbreak of COVID-19 has become a global crisis, and brought severe disruptions to societies and economies. Until now, effective therapeutics against COVID-19 are in high demand. Along with our improved understanding of the structure, function, and pathogenic process of SARS-CoV-2, many small molecules with potential anti-COVID-19 effects have been developed. So far, several antiviral strategies were explored. Besides directly inhibition of viral proteins such as RdRp and Mpro, interference of host enzymes including ACE2 and proteases, and blocking relevant immunoregulatory pathways represented by JAK/STAT, BTK, NF-κB, and NLRP3 pathways, are regarded feasible in drug development. The development of small molecules to treat COVID-19 has been achieved by several strategies, including computer-aided lead compound design and screening, natural product discovery, drug repurposing, and combination therapy. Several small molecules representative by remdesivir and paxlovid have been proved or authorized emergency use in many countries. And many candidates have entered clinical-trial stage. Nevertheless, due to the epidemiological features and variability issues of SARS-CoV-2, it is necessary to continue exploring novel strategies against COVID-19. This review discusses the current findings in the development of small molecules for COVID-19 treatment. Moreover, their detailed mechanism of action, chemical structures, and preclinical and clinical efficacies are discussed.
Collapse
Affiliation(s)
- Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xiaohua Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
34
|
de Cos M, Meliambro K, Campbell KN. Novel Treatment Paradigms: Focal Segmental Glomerulosclerosis. Kidney Int Rep 2022; 8:30-35. [PMID: 36644367 PMCID: PMC9831941 DOI: 10.1016/j.ekir.2022.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a histologic pattern of injury defined by the presence of sclerosis in some (segmental) of certain glomeruli (focal). On electron microscopy, it is characterized by a variable degree of podocyte foot process effacement and gaps in the coverage of the glomerular basement membrane. The pattern of injury occurs when podocytes, highly differentiated cells with limited regenerative capacity, are reduced in number. The heterogeneity in underlying causes of podocyte loss results in equally variable clinical phenotypes. Recent work acknowledging advances in defining the genetic and immunologic basis of disease has redefined the classification of FSGS. Unprecedented clinical trial activity and efficacy of repurposed agents presents hope for improved therapeutic options. This minireview summarizes recent advances with a focus on novel treatment paradigms in FSGS.
Collapse
Affiliation(s)
- Marina de Cos
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristin Meliambro
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirk N. Campbell
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Correspondence: Kirk N. Campbell, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.
| |
Collapse
|