51
|
Freedman BI, Limou S, Ma L, Kopp JB. APOL1-Associated Nephropathy: A Key Contributor to Racial Disparities in CKD. Am J Kidney Dis 2019; 72:S8-S16. [PMID: 30343724 DOI: 10.1053/j.ajkd.2018.06.020] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/25/2018] [Indexed: 01/09/2023]
Abstract
Genetic methodologies are improving our understanding of the pathophysiology in diverse diseases. Breakthroughs have been particularly impressive in nephrology, for which marked disparities exist in rates and etiologic classifications of end-stage kidney disease between African Americans and European Americans. Discovery of the apolipoprotein L1 gene (APOL1) association with focal segmental glomerulosclerosis, human immunodeficiency virus (HIV)-associated nephropathy, lupus nephritis, sickle cell nephropathy, and solidified glomerulosclerosis, as well as more rapid failure of transplanted kidneys from donors with APOL1 renal-risk genotypes, has improved our understanding of nondiabetic nephropathy. Environmental factors acting through natural selection in sub-Saharan African populations likely underlie this association. This article describes the discovery of chromosome 22q renal-risk variants and their worldwide distribution, reviews the epidemiology and pathology of APOL1-associated nephropathies, and explores several proposed mechanisms of kidney injury identified in cell culture and animal models. Detection of APOL1 associations with kidney diseases and delineation of injury pathways brings hope for effective treatment for these kidney diseases.
Collapse
Affiliation(s)
- Barry I Freedman
- Section on Nephrology, Department of Internal Medicine; Wake Forest School of Medicine, Winston-Salem, NC.
| | - Sophie Limou
- Centre de Recherche en Transplantation et Immunologie (CRTI) UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie et Néphrologie (ITUN), CHU Nantes, Nantes, France; Ecole Centrale de Nantes, Nantes, France
| | - Lijun Ma
- Section on Nephrology, Department of Internal Medicine; Wake Forest School of Medicine, Winston-Salem, NC
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
52
|
Lannon H, Shah SS, Dias L, Blackler D, Alper SL, Pollak MR, Friedman DJ. Apolipoprotein L1 (APOL1) risk variant toxicity depends on the haplotype background. Kidney Int 2019; 96:1303-1307. [PMID: 31611067 DOI: 10.1016/j.kint.2019.07.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/27/2019] [Accepted: 07/03/2019] [Indexed: 01/01/2023]
Abstract
The Apolipoprotein L1 (APOL1) risk variants G1 and G2 are associated with high rates of kidney disease in African Americans in genetic studies. However, our understanding of APOL1 biology has lagged far behind. Here we report that engineering G1 and G2 mutations on unnatural haplotype backgrounds instead of on the specific G1 and G2 haplotype backgrounds that occur in nature profoundly alters APOL1-mediated cytotoxicity in experimental systems. Thus, in addition to helping resolve some important controversies in the APOL1 field, our demonstration of the critical influence of haplotype background may apply more generally to the study of other genetic variants that cause or predispose to human disease.
Collapse
Affiliation(s)
- Herbert Lannon
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Shrijal S Shah
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Leny Dias
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Blackler
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Seth L Alper
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Martin R Pollak
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - David J Friedman
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
53
|
Abstract
PURPOSE OF REVIEW The purpose of this mini-review is to highlight some unresolved questions and controversies in the evolving story of apolipoprotein L1 (APOL1) nephropathy. RECENT FINDINGS We highlight studies that introduce complexity in unraveling the mechanisms whereby APOL1 risk variant alleles cause disease. These include studies which support a possible protective role for the APOL1 GO nonrisk ancestral allele, and studies which explore the initiating events that may trigger other downstream pathways mediating APOL1 cellular injury. We also review studies that reconcile the perplexing findings regarding APOL1 anionic or cationic conductance, and pH dependency, and also studies that attempt to characterize the 3-dimensional structure of APOL1 C-terminal in APOL1 variants, as well as that of the serum resistance-associated protein. We also attempt to convey new insights from in-vivo and in-vitro models, including studies that do not support the differential toxicity of APOL1 renal risk variants and recapitulate the clinical variability of individuals at genotypic risk. SUMMARY Along with major progress that had been achieved in the field of APOL1 nephropathy, controversies and enigmatic issues persist. It remains to be determined which of the pathways which have been demonstrated to mediate cell injury by ectopically expressed APOL1 risk variants in cellular and organismal models are relevant to human disease and can pave the way to potential therapy.
Collapse
|
54
|
Abstract
The apolipoprotein L1 (APOL1) gene is unique to humans and gorillas and appeared ~33 million years ago. Since the majority of the mammals do not carry APOL1, it seems to be dispensable for kidney function. APOL1 renal risk variants (RRVs; G1 and G2) are associated with the development as well as progression of chronic kidney diseases (CKDs) at higher rates in populations with African ancestry. Cellular expression of two APOL1 RRVs has been demonstrated to induce cytotoxicity, including necrosis, apoptosis, and pyroptosis, in several cell types including podocytes; mechanistically, these toxicities were attributed to lysosomal swelling, K+ depletion, mitochondrial dysfunction, autophagy blockade, protein kinase receptor activation, ubiquitin D degradation, and endoplasmic reticulum stress; notably, these effects were found to be dose dependent and occurred only in overtly APOL1 RRV-expressing cells. However, cellular protein expressions as well as circulating blood levels of APOL1 RRVs were not elevated in patients suffering from APOL1 RRV-associated CKDs. Therefore, the question arises as to whether it is gain or loss of function on the part of APOL1 RRVs contributing to kidney cell injury. The question seems to be more pertinent after the recognition of the role of APOL1 nonrisk (G0) in the transition of parietal epithelial cells and preservation of the podocyte molecular phenotype through modulation of the APOL1-miR-193a axis. With this background, the present review analyzed the available literature in terms of the known function of APOL1 nonrisk and how the loss of these functions could have contributed to two APOL1 RRV-associated CKDs.
Collapse
Affiliation(s)
- Vinod Kumar
- Institute of Molecular Medicine, Feinstein Institute for Medical Research and Department of Medicine, Zucker School of Medicine at Hofstra-Northwell, Hempstead, New York
| | - Pravin C Singhal
- Institute of Molecular Medicine, Feinstein Institute for Medical Research and Department of Medicine, Zucker School of Medicine at Hofstra-Northwell, Hempstead, New York
| |
Collapse
|
55
|
Aghajan M, Booten SL, Althage M, Hart CE, Ericsson A, Maxvall I, Ochaba J, Menschik-Lundin A, Hartleib J, Kuntz S, Gattis D, Ahlström C, Watt AT, Engelhardt JA, Monia BP, Magnone MC, Guo S. Antisense oligonucleotide treatment ameliorates IFN-γ-induced proteinuria in APOL1-transgenic mice. JCI Insight 2019; 4:126124. [PMID: 31217349 DOI: 10.1172/jci.insight.126124] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 05/16/2019] [Indexed: 12/20/2022] Open
Abstract
African Americans develop end-stage renal disease at a higher rate compared with European Americans due to 2 polymorphisms (G1 and G2 risk variants) in the apolipoprotein L1 (APOL1) gene common in people of African ancestry. Although this compelling genetic evidence provides an exciting opportunity for personalized medicine in chronic kidney disease, drug discovery efforts have been greatly hindered by the fact that APOL1 expression is lacking in rodents. Here, we describe a potentially novel physiologically relevant genomic mouse model of APOL1-associated renal disease that expresses human APOL1 from the endogenous human promoter, resulting in expression in similar tissues and at similar relative levels as humans. While naive APOL1-transgenic mice did not exhibit a renal disease phenotype, administration of IFN-γ was sufficient to robustly induce proteinuria only in APOL1 G1 mice, despite inducing kidney APOL1 expression in both G0 and G1 mice, serving as a clinically relevant "second hit." Treatment of APOL1 G1 mice with IONIS-APOL1Rx, an antisense oligonucleotide (ASO) targeting APOL1 mRNA, prior to IFN-γ challenge robustly and dose-dependently inhibited kidney and liver APOL1 expression and protected against IFN-γ-induced proteinuria, indicating that the disease-relevant cell types are sensitive to ASO treatment. Therefore, IONIS-APOL1Rx may be an effective therapeutic for APOL1 nephropathies and warrants further development.
Collapse
Affiliation(s)
| | | | - Magnus Althage
- Cardiovascular, Renal and Metabolic Diseases, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | | | - Anette Ericsson
- Cardiovascular, Renal and Metabolic Diseases, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Ingela Maxvall
- Cardiovascular, Renal and Metabolic Diseases, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | | | - Angela Menschik-Lundin
- Cardiovascular, Renal and Metabolic Diseases, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Judith Hartleib
- Cardiovascular, Renal and Metabolic Diseases, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Steven Kuntz
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | - Christine Ahlström
- Cardiovascular, Renal and Metabolic Diseases, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | | | | | | | - Maria Chiara Magnone
- Cardiovascular, Renal and Metabolic Diseases, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Shuling Guo
- Ionis Pharmaceuticals, Carlsbad, California, USA
| |
Collapse
|
56
|
Kumar V, Ayasolla K, Jha A, Mishra A, Vashistha H, Lan X, Qayyum M, Chinnapaka S, Purohit R, Mikulak J, Saleem MA, Malhotra A, Skorecki K, Singhal PC. Disrupted apolipoprotein L1-miR193a axis dedifferentiates podocytes through autophagy blockade in an APOL1 risk milieu. Am J Physiol Cell Physiol 2019; 317:C209-C225. [PMID: 31116585 DOI: 10.1152/ajpcell.00538.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We hypothesized that a functional apolipoprotein LI (APOL1)-miR193a axis (inverse relationship) preserves, but disruption alters, the podocyte molecular phenotype through the modulation of autophagy flux. Podocyte-expressing APOL1G0 (G0-podocytes) showed downregulation but podocyte-expressing APOL1G1 (G1-podocytes) and APOL1G2 (G2-podocytes) displayed enhanced miR193a expression. G0-, G1-, and G2-podocytes showed enhanced expression of light chain (LC) 3-II and beclin-1, but a disparate expression of p62 (low in wild-type but high in risk alleles). G0-podocytes showed enhanced, whereas G1- and G2-podocytes displayed decreased, phosphorylation of Unc-51-like autophagy-activating kinase (ULK)1 and class III phosphatidylinositol 3-kinase (PI3KC3). Podocytes overexpressing miR193a (miR193a-podocytes), G1, and G2 showed decreased transcription of PIK3R3 (PI3KC3's regulatory unit). Since 3-methyladenine (3-MA) enhanced miR193a expression but inhibited PIK3R3 transcription, it appears that 3-MA inhibits autophagy and induces podocyte dedifferentiation via miR193a generation. miR193a-, G1-, and G2-podocytes also showed decreased phosphorylation of mammalian target of rapamycin (mTOR) that could repress lysosome reformation. G1- and G2-podocytes showed enhanced expression of run domain beclin-1-interacting and cysteine-rich domain-containing protein (Rubicon); however, its silencing prevented their dedifferentiation. Docking, protein-protein interaction, and immunoprecipitation studies with antiautophagy-related gene (ATG)14L, anti-UV radiation resistance-associated gene (UVRAG), or Rubicon antibodies suggested the formation of ATG14L complex I and UVRAG complex II in G0-podocytes and the formation of Rubicon complex III in G1- and G2-podocytes. These findings suggest that the APOL1 risk alleles favor podocyte dedifferentiation through blockade of multiple autophagy pathways.
Collapse
Affiliation(s)
- Vinod Kumar
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Kamesh Ayasolla
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Alok Jha
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Abheepsa Mishra
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | | | - Xiqian Lan
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Maleeha Qayyum
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Sushma Chinnapaka
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Richa Purohit
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Joanna Mikulak
- Humanitas Clinical and Research Center, Rozzano, Milan , Italy
| | - Moin A Saleem
- Academic Renal Unit, University of Bristol , Bristol , United Kingdom
| | - Ashwani Malhotra
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| | - Karl Skorecki
- Technion-Israel Institute of Technology, Rambam Health Care Campus, Haifa , Israel
| | - Pravin C Singhal
- Feinstein Institute and Zucker School of Medicine at Hofstra-Northwell , Hempstead, New York
| |
Collapse
|
57
|
Abstract
PURPOSE OF REVIEW APOL1 nephropathy risk variants drive most of the excess risk of chronic kidney disease (CKD) seen in African Americans, but whether the same risk variants account for excess cardiovascular risk remains unclear. This mini-review highlights the controversies in the APOL1 cardiovascular field. RECENT FINDINGS In the past 10 years, our understanding of how APOL1 risk variants contribute to renal cytotoxicity has increased. Some of the proposed mechanisms for kidney disease are biologically plausible for cells and tissues relevant to cardiovascular disease (CVD), but cardiovascular studies published since 2014 have reported conflicting results regarding APOL1 risk variant association with cardiovascular outcomes. In the past year, several studies have also contributed conflicting results from different types of study populations. SUMMARY Heterogeneity in study population and study design has led to differing reports on the role of APOL1 nephropathy risk variants in CVD. Without consistently validated associations between these risk variants and CVD, mechanistic studies for APOL1's role in cardiovascular biology lag behind.
Collapse
|
58
|
Kumar V, Paliwal N, Ayasolla K, Vashistha H, Jha A, Chandel N, Chowdhary S, Saleem MA, Malhotra A, Chander PN, Skorecki K, Singhal PC. Disruption of APOL1-miR193a Axis Induces Disorganization of Podocyte Actin Cytoskeleton. Sci Rep 2019; 9:3582. [PMID: 30837512 PMCID: PMC6401370 DOI: 10.1038/s41598-019-39376-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/22/2018] [Indexed: 02/08/2023] Open
Abstract
APOL1-miR193a axis participates in the preservation of molecular phenotype of differentiated podocytes (DPDs). We examined the hypothesis that APOL1 (G0) preserves, but APOL1 risk alleles (G1 and G2) disrupt APOL1-miR193a axis in DPDs. DPDG0s displayed down-regulation of miR193a, but upregulation of nephrin expression. DPDG1s/G2s exhibited an increase in miR193a and down-regulation of the expression of adherens complex's constituents (CD2AP, nephrin, and dendrin). DPDG0s showed decreased Cathepsin L, enhanced dynamin expressions, and the intact actin cytoskeleton. On the contrary, DPDG1s/G2s displayed an increase in Cathepsin L, but down-regulation of dynamin expressions and disorganization of the actin cytoskeleton. APOL1 silencing enhanced miR193a and Cathepsin L, but down-regulated dynamin expressions. DPDG1s/G2s displayed nuclear import of dendrin, indicating an occurrence of destabilization of adherens complexes in APOL1 risk milieu. These findings suggest that DPDG1s and DPDG2s developed disorganized actin cytoskeleton as a consequence of disrupted APOL1-miR193a axis. Interestingly, docking and co-labeling studies suggested an interaction between APOL1 and CD2AP. APOL1G1/G1 and APOL1G1/G2 transgenic mice displayed nuclear import of dendrin indicating destabilization of adherens complexes in podocytes; moreover, these mice showed a four-fold increase in urinary albumin to creatinine ratio and development of focal segmental glomerular lesions.
Collapse
Affiliation(s)
- Vinod Kumar
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA
| | - Nitpriya Paliwal
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA
| | - Kamesh Ayasolla
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA
| | | | - Alok Jha
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA
| | - Nirupama Chandel
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA
| | - Sheetal Chowdhary
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA
| | | | - Ashwani Malhotra
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA
| | | | - Karl Skorecki
- Technion - Israel Institute of Technology, and Rambam Health Care Campus, Haifa, Israel
| | - Pravin C Singhal
- Immunology and Inflammation Center, Feinstein Institute for Medical Research and Zucker School of Medicine at Hofstra-Northwell, New York, USA.
| |
Collapse
|
59
|
Abstract
BACKGROUND An improved understanding of the pathogenesis in apolipoprotein L1 (APOL1) gene-associated chronic kidney disease (CKD) arose from observations in kidney transplantation. APOL1 genotyping could soon improve the safety of living kidney donation in individuals with recent African ancestry and alter the allocation of deceased donor kidneys. METHODS This article reviews the potential mechanisms that underlie development of APOL1-associated nephropathy. Roles for circulating APOL1 protein versus intrinsic renal expression of APOL1 are discussed, as well as the requirement for modifying genetic and/or environmental factors. RESULTS Abundant evidence supports local kidney production of APOL1 renal-risk variant protein in the development of nephropathy; this is true in both native kidney disease and after renal transplantation. Only a minority of kidneys from individuals with APOL1 high-risk genotypes will develop CKD or manifest shorter renal allograft survival after transplantation. Therefore, modifying factors that explain why only a subset of kidneys develops nephropathy remain critical to identify. It appears likely that environmental exposures, as opposed to major APOL1-second gene interactions, will prove to be stronger modifiers of the risk for nephropathy. CONCLUSIONS The evolving understanding of the pathogenesis in APOL1-associated nephropathy will identify biomarkers predicting nephropathy in individuals at high genetic risk and lead to novel therapies to prevent or slow native CKD progression and prolong survival of transplanted kidneys. In the interim, the National Institutes of Health-sponsored "APOL1 Long-term Kidney Transplantation Outcomes" Network will determine whether APOL1 genotyping in individuals with recent African ancestry improves outcomes and safety in kidney transplantation.
Collapse
Affiliation(s)
- Lijun Ma
- Department of Internal Medicine, Section on Nephrology; Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jasmin Divers
- Division of Public Health Sciences, Department of Biostatistical Sciences; Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Barry I. Freedman
- Department of Internal Medicine, Section on Nephrology; Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
60
|
Advances in molecular diagnosis and therapeutics in nephrotic syndrome and focal and segmental glomerulosclerosis. Curr Opin Nephrol Hypertens 2019; 27:194-200. [PMID: 29465426 DOI: 10.1097/mnh.0000000000000408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW The widespread adoption of next-generation sequencing by research and clinical laboratories has begun to uncover the previously unknown genetic basis of many diseases. In nephrology, one of the best examples of this is seen in focal and segmental glomerulosclerosis (FSGS) and nephrotic syndrome. We review advances made in 2017 as a result of human and molecular genetic studies as it relates to FSGS and nephrotic syndrome. RECENT FINDINGS There are more than 50 monogenic genes described in steroid-resistant nephrotic syndrome and FSGS, with seven reported in 2017. In individuals presenting with FSGS or nephrotic syndrome before or at the age of 18 years, the commonest genes in which a mutation is found continues to be limited to only a few including NPHS1 and NPHS2 based on multiple studies. For FSGS or nephrotic syndrome that presents after 18 years, mutations in COl4A3/4/5, traditionally associated with Alport syndrome, are increasingly being reported. Despite the extensive genetic heterogeneity in FSGS, there is evidence that some of these genes converge onto common pathways. There are also reports of in-vivo models exploring apolipoprotein 1 biology, variants in which account for part of the increased risk of nondiabetic kidney disease in African-Americans. Finally, genetic testing has several clinical uses including clarification of diagnosis and treatment; identification of suitable young biologic relatives for kidney donation; and preimplantation genetic diagnosis. CRISPR gene editing is currently an experimental tool only, but the recent reports of excising mutations in embryos could be a therapeutic option for individuals with any monogenic disorder in the future. SUMMARY Sequencing efforts are bringing novel variants into investigation and directing the efforts to understand how these lead to disease phenotypes. Expanding our understanding of the genetic basis of health and disease processes is the necessary first step to elaborate the repertoire of therapeutic agents available for patients with FSGS and nephrotic syndrome.
Collapse
|
61
|
An P, Kirk GD, Limou S, Binns-Roemer E, Kopp JB, Winkler CA. Impact of APOL1 Genetic Variants on HIV-1 Infection and Disease Progression. Front Immunol 2019; 10:53. [PMID: 30733721 PMCID: PMC6353846 DOI: 10.3389/fimmu.2019.00053] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/09/2019] [Indexed: 01/03/2023] Open
Abstract
Apolipoprotein L1 (APOL1) has broad innate immune functions and has been shown to restrict HIV replication in vitro by multiple mechanisms. Coding variants in APOL1 are strongly associated with HIV-associated nephropathy (HIVAN) in persons with untreated HIV infection; however, the mechanism by which APOL1 variant protein potentiates renal injury in the presence of high viral load is not resolved. Little is known about the association of APOL1 genotypes with HIV viral load, HIV acquisition, or progression to AIDS. We assessed the role of APOL1 coding variants on HIV-1 acquisition using the conditional logistic regression test, on viral load using the t-test or ANOVA, and on progression to AIDS using Cox proportional hazards models among African Americans enrolled in the ALIVE HIV natural history cohort (n = 775). APOL1 variants were not associated with susceptibility to HIV-1 acquisition by comparing genotype frequencies between HIV-1 positive and exposed or at-risk HIV-1 uninfected groups (recessive model, 12.8 vs. 12.5%, respectively; OR 1.02, 95% CI 0.62-1.70). Similar null results were observed for dominant and additive models. APOL1 variants were not associated with HIV-1 viral load or with risk of progression to AIDS [Relative hazards (RH) 1.33, 95% CI 0.30-5.89 and 0.96, 95% CI 0.49-1.88, for recessive and additive models, respectively]. In summary, we found no evidence that APOL1 variants are associated with host susceptibility to HIV-1 acquisition, set-point HIV-1 viral load or time to incident AIDS. These results suggest that APOL1 variants are unlikely to influence HIV infection or progression among individuals of African ancestry.
Collapse
Affiliation(s)
- Ping An
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Gregory D Kirk
- Departments of Epidemiology and Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Sophie Limou
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States.,CRTI UMR1064, Inserm, Université de Nantes & ITUN, CHU Nantes, Nantes, France.,Ecole Centrale de Nantes, Nantes, France
| | - Elizabeth Binns-Roemer
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, United States
| | - Cheryl A Winkler
- Molecular Genetic Epidemiology Section, Basic Science Program, Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| |
Collapse
|
62
|
Okamoto K, Rausch JW, Wakashin H, Fu Y, Chung JY, Dummer PD, Shin MK, Chandra P, Suzuki K, Shrivastav S, Rosenberg AZ, Hewitt SM, Ray PE, Noiri E, Le Grice SFJ, Hoek M, Han Z, Winkler CA, Kopp JB. APOL1 risk allele RNA contributes to renal toxicity by activating protein kinase R. Commun Biol 2018; 1:188. [PMID: 30417125 PMCID: PMC6220249 DOI: 10.1038/s42003-018-0188-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/03/2018] [Indexed: 01/09/2023] Open
Abstract
APOL1 risk alleles associate with chronic kidney disease in African Americans, but the mechanisms remain to be fully understood. We show that APOL1 risk alleles activate protein kinase R (PKR) in cultured cells and transgenic mice. This effect is preserved when a premature stop codon is introduced to APOL1 risk alleles, suggesting that APOL1 RNA but not protein is required for the effect. Podocyte expression of APOL1 risk allele RNA, but not protein, in transgenic mice induces glomerular injury and proteinuria. Structural analysis of the APOL1 RNA shows that the risk variants possess secondary structure serving as a scaffold for tandem PKR binding and activation. These findings provide a mechanism by which APOL1 variants damage podocytes and suggest novel therapeutic strategies.
Collapse
Affiliation(s)
- Koji Okamoto
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
- Department of Nephrology, Endocrinology, Hemodialysis & Apheresis, University Hospital, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 133-8655, Japan
| | - Jason W Rausch
- Reverse Transcriptase Biochemistry Section, Basic Research Program, Frederick National Laboratory for Cancer Research, 1050 Boyle Street, Frederick, MD, 21702, USA
| | - Hidefumi Wakashin
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Yulong Fu
- Children's National Health System, 111 Michigan Ave NW, Washington, DC, 20010, USA
| | - Joon-Yong Chung
- Experimental Pathology Lab, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Patrick D Dummer
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Myung K Shin
- Merck Research Laboratories, Merck and Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Preeti Chandra
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Kosuke Suzuki
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Shashi Shrivastav
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins Medical Institutions, 720 Rutland Avenue, Baltimore, MD, 21287, USA
| | - Stephen M Hewitt
- Experimental Pathology Lab, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Patricio E Ray
- Children's National Health System, 111 Michigan Ave NW, Washington, DC, 20010, USA
| | - Eisei Noiri
- Department of Nephrology, Endocrinology, Hemodialysis & Apheresis, University Hospital, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 133-8655, Japan
| | - Stuart F J Le Grice
- Reverse Transcriptase Biochemistry Section, Basic Research Program, Frederick National Laboratory for Cancer Research, 1050 Boyle Street, Frederick, MD, 21702, USA
| | - Maarten Hoek
- Merck Research Laboratories, Merck and Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Zhe Han
- Children's National Health System, 111 Michigan Ave NW, Washington, DC, 20010, USA
| | - Cheryl A Winkler
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Frederick National Laboratory, 8560 Progress Dr., Frederick, MD, 21702, USA
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| |
Collapse
|
63
|
Bruggeman LA, O'Toole JF, Sedor JR. APOL1 polymorphisms and kidney disease: loss-of-function or gain-of-function? Am J Physiol Renal Physiol 2018; 316:F1-F8. [PMID: 30332315 DOI: 10.1152/ajprenal.00426.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The mechanism that explains the association of APOL1 variants with nondiabetic kidney diseases in African Americans remains unclear. Kidney disease risk is inherited as a recessive trait, and many studies investigating the intracellular function of APOL1 have indicated the APOL1 variants G1 and G2 are associated with cytotoxicity. Whether cytotoxicity results from the absence of a protective effect conferred by the G0 allele or is induced by a deleterious effect of variant allele expression has not be conclusively established. A central issue hampering basic biology studies is the lack of model systems that authentically replicate APOL1 expression patterns. APOL1 is present in humans and a few other primates and appears to have important functions in the kidney, as the kidney is the primary target for disease associated with the genetic variance. There have been no studies to date assessing the function of untagged APOL1 protein under native expression in human or primate kidney cells, and no studies have examined the heterozygous state, a disease-free condition in humans. A second major issue is the chronic kidney disease (CKD)-associated APOL1 variants are conditional mutations, where the disease-inducing function is only evident under the appropriate environmental stimulus. In addition, it is possible there may be more than one mechanism of pathogenesis that is dependent on the nature of the stressor or other genetic variabilities. Studies addressing the function of APOL1 and how the CKD-associated APOL1 variants cause kidney disease are challenging and remain to be fully investigated under conditions that faithfully model known human genetics and physiology.
Collapse
Affiliation(s)
- Leslie A Bruggeman
- Department of Inflammation and Immunity, Cleveland Clinic , Cleveland, Ohio.,Department of Nephrology, Cleveland Clinic , Cleveland, Ohio
| | - John F O'Toole
- Department of Inflammation and Immunity, Cleveland Clinic , Cleveland, Ohio.,Department of Nephrology, Cleveland Clinic , Cleveland, Ohio
| | - John R Sedor
- Department of Inflammation and Immunity, Cleveland Clinic , Cleveland, Ohio.,Department of Nephrology, Cleveland Clinic , Cleveland, Ohio.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine , Cleveland, Ohio
| |
Collapse
|
64
|
Hochapfel F, Denk L, Maaßen C, Zaytseva Y, Rachel R, Witzgall R, Krahn MP. Electron microscopy of
Drosophila
garland cell nephrocytes: Optimal preparation, immunostaining and STEM tomography. J Cell Biochem 2018; 119:8011-8021. [DOI: 10.1002/jcb.26702] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/23/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Florian Hochapfel
- Molecular and Cellular AnatomyUniversity of RegensburgRegensburgGermany
- Medizinische Klinik und Poliklinik DUniversitätsklinikum MünsterMünsterGermany
| | - Lucia Denk
- Molecular and Cellular AnatomyUniversity of RegensburgRegensburgGermany
| | - Christine Maaßen
- Molecular and Cellular AnatomyUniversity of RegensburgRegensburgGermany
| | - Yulia Zaytseva
- Molecular and Cellular AnatomyUniversity of RegensburgRegensburgGermany
| | - Reinhard Rachel
- Molecular and Cellular AnatomyUniversity of RegensburgRegensburgGermany
| | - Ralph Witzgall
- Molecular and Cellular AnatomyUniversity of RegensburgRegensburgGermany
| | - Michael P. Krahn
- Molecular and Cellular AnatomyUniversity of RegensburgRegensburgGermany
- Medizinische Klinik und Poliklinik DUniversitätsklinikum MünsterMünsterGermany
| |
Collapse
|
65
|
Cheatham AM, Davis SE, Khatua AK, Popik W. Blocking the 5' splice site of exon 4 by a morpholino oligomer triggers APOL1 protein isoform switch. Sci Rep 2018; 8:8739. [PMID: 29880816 PMCID: PMC5992166 DOI: 10.1038/s41598-018-27104-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/21/2018] [Indexed: 12/16/2022] Open
Abstract
APOL1 risk alleles G1 or G2 are associated with a kidney disease phenotype exclusively in people of recent African ancestry. Here we show that exon 4 encoding a part of the APOL1 signal peptide is constitutively spliced in major APOL1 transcripts expressed in kidney glomerular and tubular cells. We demonstrate that constitutive splicing of exon 4 results from a suboptimal hnRNP A1 binding motif found in exon 4. Accordingly, a robust binding of hnRNP A1 protein to a consensus hnRNP A1 cis-acting element in exon 4 results in almost complete exclusion of exon 4 from the APOL1 minigene transcripts. Blocking the 5' splice site at the exon 4/intron boundary with a specific antisense morpholino oligonucleotide excludes exon 4 from the splicing pattern of endogenous APOL1 transcripts. These transcripts are fully functional and produce APOL1 protein isoform that is not normally detectable in podocytes. Together with our previous data showing no cytotoxicity of overexpressed APOL1 isoform lacking exon 4, we propose that morpholino-induced APOL1 isoform switch may provide a new tool to identify in vivo molecular mechanism(s) by which risk alleles promote or mediate the kidney disease phenotype.
Collapse
Affiliation(s)
- Amber M Cheatham
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, 1005 D. B. Todd Blvd, Nashville, TN, 37028, USA
| | - Shamara E Davis
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, 1005 D. B. Todd Blvd, Nashville, TN, 37028, USA
| | - Atanu K Khatua
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, 1005 D. B. Todd Blvd, Nashville, TN, 37028, USA
| | - Waldemar Popik
- Meharry Medical College, Center for AIDS Health Disparities Research, Department of Microbiology and Immunology, 1005 D. B. Todd Blvd, Nashville, TN, 37028, USA.
- Department of Internal Medicine, 1005 D. B. Todd Blvd, Nashville, TN, 37028, USA.
| |
Collapse
|
66
|
APOL1: The Balance Imposed by Infection, Selection, and Kidney Disease. Trends Mol Med 2018; 24:682-695. [PMID: 29886044 DOI: 10.1016/j.molmed.2018.05.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) affects millions of people and constitutes a major health and financial burden worldwide. People of African descent are at an increased risk of developing kidney disease, which is mostly explained by two variants in the Apolipoprotein L1 (APOL1) gene that are found only in people of west African origin. It is hypothesized that these variants were genetically selected due to the protection they afford against African sleeping sickness, caused by the parasite Trypanosoma brucei. Targeting mutant APOL1 could have substantial therapeutic potential for treating kidney disease. In this review, we will describe the intriguing interplay between microbiology, genetics, and kidney disease as revealed in APOL1-associated kidney disease, discuss APOL1-induced cytotoxicity and its therapeutic implications.
Collapse
|
67
|
Palau L, Menez S, Rodriguez-Sanchez J, Novick T, Delsante M, McMahon BA, Atta MG. HIV-associated nephropathy: links, risks and management. HIV AIDS (Auckl) 2018; 10:73-81. [PMID: 29872351 PMCID: PMC5975615 DOI: 10.2147/hiv.s141978] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Despite the decreased incidence of human immunodeficiency virus (HIV)-associated nephropathy due to the widespread use of combined active antiretroviral therapy, it remains one of the leading causes of end-stage renal disease (ESRD) in HIV-1 seropositive patients. Patients usually present with low CD4 count, high viral load and heavy proteinuria, with the pathologic findings of collapsing focal segmental glomerulosclerosis. Increased susceptibility exists in individuals with African descent, largely due to polymorphism in APOL1 gene. Other clinical risk factors include high viral load and low CD4 count. Advanced kidney disease and nephrotic range proteinuria have been associated with progression to ESRD. Improvement in kidney function has been observed after initiation of combined active antiretroviral therapy. Other treatment options, when clinically indicated, are inhibition of the renin-angiotensin system and corticosteroids. Further routine management approaches for patients with chronic kidney disease should be implemented. In patients with progression to ESRD, kidney transplant should be pursued, provided that viral load control is adequate. Screening for the presence of kidney disease upon detection of HIV-1 seropositivity in high-risk populations is recommended.
Collapse
Affiliation(s)
- Laura Palau
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Steven Menez
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Tessa Novick
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Marco Delsante
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Blaithin A McMahon
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mohamed G Atta
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
68
|
Gonçalves S, Patat J, Guida MC, Lachaussée N, Arrondel C, Helmstädter M, Boyer O, Gribouval O, Gubler MC, Mollet G, Rio M, Charbit M, Bole-Feysot C, Nitschke P, Huber TB, Wheeler PG, Haynes D, Juusola J, Billette de Villemeur T, Nava C, Afenjar A, Keren B, Bodmer R, Antignac C, Simons M. A homozygous KAT2B variant modulates the clinical phenotype of ADD3 deficiency in humans and flies. PLoS Genet 2018; 14:e1007386. [PMID: 29768408 PMCID: PMC5973622 DOI: 10.1371/journal.pgen.1007386] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 05/29/2018] [Accepted: 04/30/2018] [Indexed: 12/25/2022] Open
Abstract
Recent evidence suggests that the presence of more than one pathogenic mutation in a single patient is more common than previously anticipated. One of the challenges hereby is to dissect the contribution of each gene mutation, for which animal models such as Drosophila can provide a valuable aid. Here, we identified three families with mutations in ADD3, encoding for adducin-γ, with intellectual disability, microcephaly, cataracts and skeletal defects. In one of the families with additional cardiomyopathy and steroid-resistant nephrotic syndrome (SRNS), we found a homozygous variant in KAT2B, encoding the lysine acetyltransferase 2B, with impact on KAT2B protein levels in patient fibroblasts, suggesting that this second mutation might contribute to the increased disease spectrum. In order to define the contribution of ADD3 and KAT2B mutations for the patient phenotype, we performed functional experiments in the Drosophila model. We found that both mutations were unable to fully rescue the viability of the respective null mutants of the Drosophila homologs, hts and Gcn5, suggesting that they are indeed pathogenic in flies. While the KAT2B/Gcn5 mutation additionally showed a significantly reduced ability to rescue morphological and functional defects of cardiomyocytes and nephrocytes (podocyte-like cells), this was not the case for the ADD3 mutant rescue. Yet, the simultaneous knockdown of KAT2B and ADD3 synergistically impaired kidney and heart function in flies as well as the adhesion and migration capacity of cultured human podocytes, indicating that mutations in both genes may be required for the full clinical manifestation. Altogether, our studies describe the expansion of the phenotypic spectrum in ADD3 deficiency associated with a homozygous likely pathogenic KAT2B variant and thereby identify KAT2B as a susceptibility gene for kidney and heart disease in ADD3-associated disorders.
Collapse
Affiliation(s)
- Sara Gonçalves
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Laboratory of Epithelial Biology and Disease, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Julie Patat
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Maria Clara Guida
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, United States of America
| | - Noelle Lachaussée
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Christelle Arrondel
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Martin Helmstädter
- Department of Medicine IV, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Olivia Boyer
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
- Department of Pediatric Nephrology, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Necker-Enfants Malades, Assistance Publique—Hôpitaux de Paris (AP-HP), Paris, France
| | - Olivier Gribouval
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Marie-Claire Gubler
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Geraldine Mollet
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Marlène Rio
- Department of Genetics, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Marina Charbit
- Department of Pediatric Nephrology, Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Hôpital Necker-Enfants Malades, Assistance Publique—Hôpitaux de Paris (AP-HP), Paris, France
| | | | - Patrick Nitschke
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Tobias B. Huber
- Department of Medicine IV, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- BIOSS Center for Biological Signalling Studies and Center for Systems Biology (ZBSA), Albert-Ludwigs-University, Freiburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patricia G. Wheeler
- Division of Genetics, Arnold Palmer Hospital for Children, Orlando Health, Orlando, FL, United States of America
| | - Devon Haynes
- Division of Genetics, Arnold Palmer Hospital for Children, Orlando Health, Orlando, FL, United States of America
| | - Jane Juusola
- GeneDx, Inc, Gaithersburg, MD, United States of America
| | - Thierry Billette de Villemeur
- Sorbonne Université, UPMC, GRC ConCer-LD and AP-HP, Hôpital Trousseau, Service de Neuropédiatrie—Pathologie du développement, Paris, France
- Centre de référence des déficits intellectuels de causes rares, Inserm U 1141, Paris, France
| | - Caroline Nava
- Sorbonne Universités, UPMC Univ Paris 06, Inserm U1127, CNRS UMR 7225, Institut du Cerveau et de la Moèlle Épinière (ICM), Paris, France
- AP-HP, GH Pitié-Salpêtrière, Department of Genetics, Unit of Developmental Genomics, Paris, France
| | - Alexandra Afenjar
- AP-HP, Hôpital Trousseau, Centre de référence des malformations et maladies congénitales du cervelet, Département de génétique et embryologie médicale, Paris, France
| | - Boris Keren
- AP-HP, GH Pitié-Salpêtrière, Department of Genetics, Unit of Developmental Genomics, Paris, France
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, United States of America
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
- Department of Genetics, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
- * E-mail: (CA); (MS)
| | - Matias Simons
- Laboratory of Epithelial Biology and Disease, Institut National de la Santé et de la Recherche Médicale (Inserm) UMR1163, Imagine Institute, Paris, France
- Université Paris Descartes—Sorbonne Paris Cité, Imagine Institute, Paris, France
- * E-mail: (CA); (MS)
| |
Collapse
|
69
|
Reidy KJ, Hjorten R, Parekh RS. Genetic risk of APOL1 and kidney disease in children and young adults of African ancestry. Curr Opin Pediatr 2018; 30:252-259. [PMID: 29406442 PMCID: PMC6002812 DOI: 10.1097/mop.0000000000000603] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Understanding the genetic risk of APOL1 in children and young adults is important given the lifetime risk of hypertension and kidney disease among children of African descent. We review recent epidemiologic and biologic findings on the effects of APOL1 and kidney disease. RECENT FINDINGS APOL1 in children and young adults is associated with hypertension, albuminuria and more rapid decline in kidney function and progression to end-stage kidney disease, especially among those with glomerular causes of kidney disease, and those affected by sickle cell disease or HIV. There are conflicting data on the APOL1 association with cardiovascular disease in children and young adults. APOL1 functions as part of the innate immune system. Podocyte expression of APOL1 likely contributes to the development of kidney disease. In cell culture and model organisms, APOL1 expression disrupts autophagic and ion flux, leads to defects in mitochondrial respiration and induces cell death. SUMMARY APOL1 explains almost 70% of the excess risk of kidney disease in those of African descent, and is common in children with glomerular disease. An evolving understanding of the pathogenesis of APOL1-mediated kidney damage may aid in personalized medicine approaches to APOL1 attributable kidney disease.
Collapse
Affiliation(s)
- Kimberly J Reidy
- Children’s Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rebecca Hjorten
- Children’s Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center
| | - Rulan S. Parekh
- Departments of Pediatrics and Medicine, Hospital for Sick Children, University Health Network and University of Toronto, Toronto, Canada
| |
Collapse
|
70
|
Kidney disease in the setting of HIV infection: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int 2018; 93:545-559. [PMID: 29398134 DOI: 10.1016/j.kint.2017.11.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/23/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022]
Abstract
HIV-positive individuals are at increased risk for kidney disease, including HIV-associated nephropathy, noncollapsing focal segmental glomerulosclerosis, immune-complex kidney disease, and comorbid kidney disease, as well as kidney injury resulting from prolonged exposure to antiretroviral therapy or from opportunistic infections. Clinical guidelines for kidney disease prevention and treatment in HIV-positive individuals are largely extrapolated from studies in the general population, and do not fully incorporate existing knowledge of the unique HIV-related pathways and genetic factors that contribute to the risk of kidney disease in this population. We convened an international panel of experts in nephrology, renal pathology, and infectious diseases to define the pathology of kidney disease in the setting of HIV infection; describe the role of genetics in the natural history, diagnosis, and treatment of kidney disease in HIV-positive individuals; characterize the renal risk-benefit of antiretroviral therapy for HIV treatment and prevention; and define best practices for the prevention and management of kidney disease in HIV-positive individuals.
Collapse
|
71
|
Cohen SD, Kopp JB, Kimmel PL. Kidney Diseases Associated with Human Immunodeficiency Virus Infection. N Engl J Med 2017; 377:2363-2374. [PMID: 29236630 DOI: 10.1056/nejmra1508467] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Scott D Cohen
- From the Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University, Washington, DC (S.D.C., P.L.K.); and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.B.K., P.L.K.)
| | - Jeffrey B Kopp
- From the Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University, Washington, DC (S.D.C., P.L.K.); and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.B.K., P.L.K.)
| | - Paul L Kimmel
- From the Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University, Washington, DC (S.D.C., P.L.K.); and the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.B.K., P.L.K.)
| |
Collapse
|
72
|
O'Toole JF, Schilling W, Kunze D, Madhavan SM, Konieczkowski M, Gu Y, Luo L, Wu Z, Bruggeman LA, Sedor JR. ApoL1 Overexpression Drives Variant-Independent Cytotoxicity. J Am Soc Nephrol 2017; 29:869-879. [PMID: 29180397 DOI: 10.1681/asn.2016121322] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 10/31/2017] [Indexed: 12/16/2022] Open
Abstract
Coding variants in the APOL1 gene are associated with kidney diseases in African ancestral populations; yet, the underlying biologic mechanisms remain uncertain. Variant-dependent autophagic and cytotoxic cell death have been proposed as pathogenic pathways mediating kidney injury. To examine this possibility, we conditionally expressed APOL1-G0 (reference), -G1, and -G2 (variants) using a tetracycline-regulated system in HEK293 cells. Autophagy was monitored biochemically and cell death was measured using multiple assays. We measured intracellular Na+ and K+ content with atomic absorption spectroscopy and APOL1-dependent currents with whole-cell patch clamping. Neither reference nor variant APOL1s induced autophagy. At high expression levels, APOL1-G0, -G1, and -G2 inserted into the plasma membrane and formed pH-sensitive cation channels, causing collapse of cellular Na+ and K+ gradients, phosphorylation of p38 mitogen-activated protein kinase, and cell death, without variant-dependent differences. APOL1-G0 and -G2 exhibited similar channel properties in whole-cell patch clamp experiments. At low expression levels, neither reference nor variant APOL1s localized on the plasma membrane, Na+ and K+ gradients were maintained, and cells remained viable. Our results indicate that APOL1-mediated pore formation is critical for the trypanolytic activity of APOL1 and drives APOL1-mediated cytotoxicity in overexpression systems. The absence of cytotoxicity at physiologic expression levels suggests variant-dependent intracellular K+ loss and cytotoxicity does not drive kidney disease progression.
Collapse
Affiliation(s)
- John F O'Toole
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio; and
| | - William Schilling
- Rammelkamp Center, MetroHealth System.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | | | | | - Yaping Gu
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Liping Luo
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Zhenzhen Wu
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Leslie A Bruggeman
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio; and
| | - John R Sedor
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio; and.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
73
|
Abstract
Chronic kidney disease (CKD) is defined by persistent urine abnormalities, structural abnormalities or impaired excretory renal function suggestive of a loss of functional nephrons. The majority of patients with CKD are at risk of accelerated cardiovascular disease and death. For those who progress to end-stage renal disease, the limited accessibility to renal replacement therapy is a problem in many parts of the world. Risk factors for the development and progression of CKD include low nephron number at birth, nephron loss due to increasing age and acute or chronic kidney injuries caused by toxic exposures or diseases (for example, obesity and type 2 diabetes mellitus). The management of patients with CKD is focused on early detection or prevention, treatment of the underlying cause (if possible) to curb progression and attention to secondary processes that contribute to ongoing nephron loss. Blood pressure control, inhibition of the renin-angiotensin system and disease-specific interventions are the cornerstones of therapy. CKD complications such as anaemia, metabolic acidosis and secondary hyperparathyroidism affect cardiovascular health and quality of life, and require diagnosis and treatment.
Collapse
|
74
|
O'Toole JF, Bruggeman LA, Sedor JR. APOL1 and Proteinuria in the AASK: Unraveling the Pathobiology of APOL1. Clin J Am Soc Nephrol 2017; 12:1723-1725. [PMID: 29051142 PMCID: PMC5672982 DOI: 10.2215/cjn.10680917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- John F O'Toole
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute and
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Leslie A Bruggeman
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute and
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - John R Sedor
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute and
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
75
|
Kruzel-Davila E, Wasser WG, Skorecki K. APOL1 Nephropathy: A Population Genetics and Evolutionary Medicine Detective Story. Semin Nephrol 2017; 37:490-507. [PMID: 29110756 DOI: 10.1016/j.semnephrol.2017.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Common DNA sequence variants rarely have a high-risk association with a common disease. When such associations do occur, evolutionary forces must be sought, such as in the association of apolipoprotein L1 (APOL1) gene risk variants with nondiabetic kidney diseases in populations of African ancestry. The variants originated in West Africa and provided pathogenic resistance in the heterozygous state that led to high allele frequencies owing to an adaptive evolutionary selective sweep. However, the homozygous state is disadvantageous and is associated with a markedly increased risk of a spectrum of kidney diseases encompassing hypertension-attributed kidney disease, focal segmental glomerulosclerosis, human immunodeficiency virus nephropathy, sickle cell nephropathy, and progressive lupus nephritis. This scientific success story emerged with the help of the tools developed over the past 2 decades in human genome sequencing and population genomic databases. In this introductory article to a timely issue dedicated to illuminating progress in this area, we describe this unique population genetics and evolutionary medicine detective story. We emphasize the paradox of the inheritance mode, the missing heritability, and unresolved associations, including cardiovascular risk and diabetic nephropathy. We also highlight how genetic epidemiology elucidates mechanisms and how the principles of evolution can be used to unravel conserved pathways affected by APOL1 that may lead to novel therapies. The APOL1 gene provides a compelling example of a common variant association with common forms of nondiabetic kidney disease occurring in a continental population isolate with subsequent global admixture. Scientific collaboration using multiple experimental model systems and approaches should further clarify pathomechanisms further, leading to novel therapies.
Collapse
Affiliation(s)
| | - Walter G Wasser
- Department of Nephrology, Rambam Health Care Campus, Haifa, Israel; Department of Nephrology, Mayanei HaYeshua Medical Center, Bnei Brak, Israel
| | - Karl Skorecki
- Department of Nephrology, Rambam Health Care Campus, Haifa, Israel; Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine and Research Institute Technion-Israel Institute of Technology, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
76
|
O’Toole JF, Bruggeman LA, Sedor JR. A New Mouse Model of APOL1 -Associated Kidney Diseases: When Traffic Gets Snarled, the Podocyte Suffers. Am J Kidney Dis 2017; 70:460-463. [DOI: 10.1053/j.ajkd.2017.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023]
|
77
|
Bruno J, Pozzi N, Oliva J, Edwards JC. Apolipoprotein L1 confers pH-switchable ion permeability to phospholipid vesicles. J Biol Chem 2017; 292:18344-18353. [PMID: 28918394 DOI: 10.1074/jbc.m117.813444] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/08/2017] [Indexed: 01/13/2023] Open
Abstract
Apolipoprotein L1 (ApoL1) is a human serum protein conferring resistance to African trypanosomes, and certain ApoL1 variants increase susceptibility to some progressive kidney diseases. ApoL1 has been hypothesized to function like a pore-forming colicin and has been reported to have permeability effects on both intracellular and plasma membranes. Here, to gain insight into how ApoL1 may function in vivo, we used vesicle-based ion permeability, direct membrane association, and intrinsic fluorescence to study the activities of purified recombinant ApoL1. We found that ApoL1 confers chloride-selective permeability to preformed phospholipid vesicles and that this selectivity is strongly pH-sensitive, with maximal activity at pH 5 and little activity above pH 7. When ApoL1 and lipid were allowed to interact at low pH and were then brought to neutral pH, chloride permeability was suppressed, and potassium permeability was activated. Both chloride and potassium permeability linearly correlated with the mass of ApoL1 in the reaction mixture, and both exhibited lipid selectivity, requiring the presence of negatively charged lipids for activity. Potassium, but not chloride, permease activity required the presence of calcium ions in both the association and activation steps. Direct assessment of ApoL1-lipid associations confirmed that ApoL1 stably associates with phospholipid vesicles, requiring low pH and the presence of negatively charged phospholipids for maximal binding. Intrinsic fluorescence of ApoL1 supported the presence of a significant structural transition when ApoL1 is mixed with lipids at low pH. This pH-switchable ion-selective permeability may explain the different effects of ApoL1 reported in intracellular and plasma membrane environments.
Collapse
Affiliation(s)
| | - Nicola Pozzi
- Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri 63110
| | - Jonathan Oliva
- Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri 63110
| | | |
Collapse
|
78
|
Hayek SS, Koh KH, Grams ME, Wei C, Ko YA, Li J, Samelko B, Lee H, Dande RR, Lee HW, Hahm E, Peev V, Tracy M, Tardi NJ, Gupta V, Altintas MM, Garborcauskas G, Stojanovic N, Winkler CA, Lipkowitz MS, Tin A, Inker LA, Levey AS, Zeier M, Freedman BI, Kopp JB, Skorecki K, Coresh J, Quyyumi AA, Sever S, Reiser J. A tripartite complex of suPAR, APOL1 risk variants and α vβ 3 integrin on podocytes mediates chronic kidney disease. Nat Med 2017; 23:945-953. [PMID: 28650456 PMCID: PMC6019326 DOI: 10.1038/nm.4362] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 06/01/2017] [Indexed: 12/17/2022]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) independently predicts chronic kidney disease (CKD) incidence and progression. Apolipoprotein L1 (APOL1) gene variants G1 and G2, but not the reference allele (G0), are associated with an increased risk of CKD in individuals of recent African ancestry. Here we show in two large, unrelated cohorts that decline in kidney function associated with APOL1 risk variants was dependent on plasma suPAR levels: APOL1-related risk was attenuated in patients with lower suPAR, and strengthened in those with higher suPAR levels. Mechanistically, surface plasmon resonance studies identified high-affinity interactions between suPAR, APOL1 and αvβ3 integrin, whereby APOL1 protein variants G1 and G2 exhibited higher affinity for suPAR-activated avb3 integrin than APOL1 G0. APOL1 G1 or G2 augments αvβ3 integrin activation and causes proteinuria in mice in a suPAR-dependent manner. The synergy of circulating factor suPAR and APOL1 G1 or G2 on αvβ3 integrin activation is a mechanism for CKD.
Collapse
Affiliation(s)
- Salim S Hayek
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kwi Hye Koh
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Morgan E Grams
- Welch Center for Prevention and Johns Hopkins Bloomberg School of Public Health, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Changli Wei
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia, USA
| | - Jing Li
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Beata Samelko
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Hyun Lee
- Center for Biomolecular Science and Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ranadheer R Dande
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Ha Won Lee
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Eunsil Hahm
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Vasil Peev
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Melissa Tracy
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Nicholas J Tardi
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Vineet Gupta
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Mehmet M Altintas
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Garrett Garborcauskas
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Nikolina Stojanovic
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Cheryl A Winkler
- Molecular Genetic Epidemiology Section, Basic Research Laboratory, Basic Science Program, NCI, Leidos Biomedical Research, Frederick National Laboratory, Frederick, Maryland, USA
| | - Michael S Lipkowitz
- Division of Nephrology and Hypertension, Georgetown University Medical Center, Washington, DC, USA
| | - Adrienne Tin
- Welch Center for Prevention and Johns Hopkins Bloomberg School of Public Health, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Martin Zeier
- Division of Nephrology, Ruprecht Karls University, Heidelberg, Germany
| | - Barry I Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Karl Skorecki
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Rambam Health Care Campus, Haifa, Israel
| | - Josef Coresh
- Welch Center for Prevention and Johns Hopkins Bloomberg School of Public Health, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sanja Sever
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Jochen Reiser
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
79
|
Madhavan SM, O'Toole JF, Konieczkowski M, Barisoni L, Thomas DB, Ganesan S, Bruggeman LA, Buck M, Sedor JR. APOL1 variants change C-terminal conformational dynamics and binding to SNARE protein VAMP8. JCI Insight 2017; 2:92581. [PMID: 28724794 DOI: 10.1172/jci.insight.92581] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
APOL1 variants in African populations mediate resistance to trypanosomal infection but increase risk for kidney diseases through unknown mechanisms. APOL1 is expressed in glomerular podocytes and does not vary with underlying kidney disease diagnoses or APOL1 genotypes, suggesting that the kidney disease-associated variants dysregulate its function rather than its localization or abundance. Structural homology searches identified vesicle-associated membrane protein 8 (VAMP8) as an APOL1 protein interactor. VAMP8 colocalizes with APOL1 in the podocyte, and the APOL1:VAMP8 interaction was confirmed biochemically and with surface plasmon resonance. APOL1 variants attenuate this interaction. Computational modeling of APOL1's 3-dimensional structure, followed by molecular dynamics simulations, revealed increased motion of the C-terminal domain of reference APOL1 compared with either variant, suggesting that the variants stabilize a closed or autoinhibited state that diminishes protein interactions with VAMP8. Changes in ellipticity with increasing urea concentrations, as assessed by circular dichroism spectroscopy, showed higher conformational stability of the C-terminal helix of the variants compared with the reference protein. These results suggest that reference APOL1 interacts with VAMP8-coated vesicles, a process attenuated by variant-induced reduction in local dynamics of the C-terminal. Disordered vesicular trafficking in the podocyte may cause injury and progressive chronic kidney diseases in susceptible African Americans subjects.
Collapse
Affiliation(s)
- Sethu M Madhavan
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth System Campus, and
| | - John F O'Toole
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth System Campus, and
| | - Martha Konieczkowski
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth System Campus, and
| | - Laura Barisoni
- Department of Pathology, University of Miami, Miami, Florida, USA
| | - David B Thomas
- Department of Pathology, University of Miami, Miami, Florida, USA
| | | | - Leslie A Bruggeman
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth System Campus, and
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - John R Sedor
- Department of Medicine and Rammelkamp Center for Education and Research, MetroHealth System Campus, and.,Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
80
|
Granado D, Müller D, Krausel V, Kruzel-Davila E, Schuberth C, Eschborn M, Wedlich-Söldner R, Skorecki K, Pavenstädt H, Michgehl U, Weide T. Intracellular APOL1 Risk Variants Cause Cytotoxicity Accompanied by Energy Depletion. J Am Soc Nephrol 2017; 28:3227-3238. [PMID: 28696248 DOI: 10.1681/asn.2016111220] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/24/2017] [Indexed: 12/17/2022] Open
Abstract
Population genetic approaches have uncovered a strong association between kidney diseases and two sequence variants of the APOL1 gene, called APOL1 risk variant G1 and variant G2, compared with the nonrisk G0 allele. However, the mechanism whereby these variants lead to disease manifestation and, in particular, whether this involves an intracellular or extracellular pool of APOL1 remains unclear. Herein, we show a predominantly intracellular localization of APOL1 G0 and the renal risk variants, which localized to membranes of the endoplasmic reticulum in podocyte cell lines. This localization did not depend on the N-terminal signal peptide that mediates APOL1 secretion into the circulation. Additionally, a fraction of these proteins localized to structures surrounding mitochondria. In vitro overexpression of G1 or G2 lacking the signal peptide inhibited cell viability, triggered phosphorylation of stress-induced kinases, increased the phosphorylation of AMP-activated protein kinase, reduced intracellular potassium levels, and reduced mitochondrial respiration rates. These findings indicate that functions at intracellular membranes, specifically those of the endoplasmic reticulum and mitochondria, are crucial factors in APOL1 renal risk variant-mediated cell injury.
Collapse
Affiliation(s)
| | | | | | | | - Christian Schuberth
- Institute of Cell Dynamics and Imaging, and Cells in Motion (CiM) Cluster of Excellence (EXC1003), University of Münster, Münster, Germany; and
| | - Melanie Eschborn
- Department of Neurology, University Hospital of Münster, Münster, Germany
| | - Roland Wedlich-Söldner
- Institute of Cell Dynamics and Imaging, and Cells in Motion (CiM) Cluster of Excellence (EXC1003), University of Münster, Münster, Germany; and
| | - Karl Skorecki
- Department of Nephrology, Rambam Health Care Campus, Haifa, Israel.,Departments of Genetics and Developmental Biology, Rappaport Faculty of Medicine and Research Institute, Technion, Israel Institute of Technology, Haifa, Israel
| | | | | | | |
Collapse
|
81
|
Helmstädter M, Simons M. Using Drosophila nephrocytes in genetic kidney disease. Cell Tissue Res 2017; 369:119-126. [PMID: 28401308 DOI: 10.1007/s00441-017-2606-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/05/2017] [Indexed: 01/01/2023]
Abstract
Renal diseases are a growing health burden, and innovative models to study their pathomechanisms are greatly needed. Here, we highlight how the fruit fly Drosophila melanogaster can be used to model kidney diseases. We focus on the nephrocyte that has recently been shown to exhibit podocyte and proximal tubular cell features. These cells can be manipulated with precise genetic tools to dissect filtration and reabsorption mechanisms. Thus, they represent a novel and easy-to-use alternative in experimental nephrology.
Collapse
Affiliation(s)
- Martin Helmstädter
- Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Matias Simons
- Imagine Institute, Paris Descartes University-Sorbonne Paris Cité, 75015, Paris, France. .,Institut Imagine, 24 Boulevard du Montparnasse, Paris, France.
| |
Collapse
|
82
|
A mouse recapitulating APOL1-associated kidney disease. Nat Med 2017; 23:411-412. [PMID: 28388606 DOI: 10.1038/nm.4318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
83
|
Abstract
Ever since the discovery of the major histocompatibility complex, scientific and clinical understanding in the field of transplantation has been advanced through genetic and genomic studies. Candidate-gene approaches and recent genome-wide association studies (GWAS) have enabled a deeper understanding of the complex interplay of the donor-recipient interactions that lead to transplant tolerance or rejection. Genetic analysis in transplantation, when linked to demographic and clinical outcomes, has the potential to drive personalized medicine by enabling individualized risk stratification and immunosuppression through the identification of variants associated with immune-mediated complications, post-transplant disease or alterations in drug-metabolizing genes.
Collapse
Affiliation(s)
- Joshua Y C Yang
- Division of Transplant Surgery, University of California San Francisco, 513 Parnassus Avenue, San Francisco, California 94143, USA
| | - Minnie M Sarwal
- Division of Transplant Surgery, University of California San Francisco, 513 Parnassus Avenue, San Francisco, California 94143, USA
| |
Collapse
|
84
|
Bruggeman LA, O'Toole JF, Sedor JR. Identifying the Intracellular Function of APOL1. J Am Soc Nephrol 2017; 28:1008-1011. [PMID: 28196842 DOI: 10.1681/asn.2016111262] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
| | | | - John R Sedor
- Department of Medicine, MetroHealth Medical Center and.,Department of Physiology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
85
|
Helmstädter M, Huber TB, Hermle T. Using the Drosophila Nephrocyte to Model Podocyte Function and Disease. Front Pediatr 2017; 5:262. [PMID: 29270398 PMCID: PMC5725439 DOI: 10.3389/fped.2017.00262] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/24/2017] [Indexed: 12/19/2022] Open
Abstract
Glomerular disorders are a major cause of end-stage renal disease and effective therapies are often lacking. Nephrocytes are considered to be part of the Drosophila excretory system and form slit diaphragms across cellular membrane invaginations. Nehphrocytes have been shown to share functional, morphological, and molecular features with podocytes, which form the glomerular filter in vertebrates. Here, we report the progress and the evolving tool-set of this model system. Combining a functional, accessible slit diaphragm with the power of the genetic tool-kit in Drosophila, the nephrocyte has the potential to greatly advance our understanding of the glomerular filtration barrier in health and disease.
Collapse
Affiliation(s)
- Martin Helmstädter
- Renal Division, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Hermle
- Renal Division, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
86
|
|