1
|
Wang QS, Huang J, Chan L, Haste N, Olsson N, Gaun A, McAllister F, Madhireddy D, Baruch A, Melamud E, Baryshnikova A. Platform-dependent effects of genetic variants on plasma APOL1 and their implications for kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635763. [PMID: 39975113 PMCID: PMC11838367 DOI: 10.1101/2025.01.30.635763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Mutations in apolipoprotein L1 (APOL1) are strongly associated with an increased risk of kidney disease in individuals of African ancestry, yet the underlying mechanisms remain largely unknown. Plasma proteomics provides opportunities to elucidate mechanisms of disease by studying the effects of disease-associated variants on circulating protein levels. Here, we examine the genetic drivers of circulating APOL1 in individuals of African and European ancestry from four independent cohorts (UK Biobank, AASK, deCODE and Health ABC) employing three proteomic technologies (Olink, SomaLogic and mass spectrometry). We find that disease-associated APOL1 G1 and G2 variants are strong pQTLs for plasma APOL1 in Olink and SomaLogic, but the direction of their effects depends on the proteomic platform. We identify an additional APOL1 missense variant (rs2239785), common in Europeans, exhibiting the same platform-dependent directional discrepancy. Similarly, variants in the kallikrein-kinin pathway ( KLKB1 , F12 , KNG1 ) and their genetic interactions exhibit strong trans -pQTL effects for APOL1 measured by Olink, but not SomaLogic. To explain these discrepancies, we propose a model in which APOL1 mutations and the kallikrein-kinin pathway influence the relative abundance of two distinct APOL1 forms, corresponding to APOL1 bound to trypanolytic factors 1 and 2, which are differentially recognized by different proteomic platforms. We hypothesize that this shift in relative abundance of APOL1 forms may contribute to the development of kidney disease.
Collapse
|
2
|
Ilatovskaya DV, Behr A, Staruschenko A, Hall G, Palygin O. Mechanistic Insights Into Redox Damage of the Podocyte in Hypertension. Hypertension 2025; 82:14-25. [PMID: 39534957 PMCID: PMC11655258 DOI: 10.1161/hypertensionaha.124.22068] [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] [Indexed: 11/16/2024]
Abstract
Podocytes are specialized cells within the glomerular filtration barrier, which are crucial for maintaining glomerular structural integrity and convective ultrafiltration. Podocytes exhibit a unique arborized morphology with foot processes interfacing by slit diaphragms, ladder-like, multimolecular sieves, which provide size and charge selectivity for ultrafiltration and transmembrane signaling. Podocyte dysfunction, resulting from oxidative stress, dysregulated prosurvival signaling, or structural damage, can drive the development of proteinuria and glomerulosclerosis in hypertensive nephropathy. Functionally, podocyte injury leads to actin cytoskeleton rearrangements, foot process effacement, dysregulated slit diaphragm protein expression, and impaired ultrafiltration. Notably, the renin-angiotensin system plays a pivotal role in podocyte function, with beneficial AT2R (angiotensin receptor 2)-mediated nitric oxide (NO) signaling to counteract AT1R (angiotensin receptor 1)-driven calcium (Ca2+) influx and oxidative stress. Disruption of this balance contributes significantly to podocyte dysfunction and drives albuminuria, a marker of kidney damage and overall disease progression. Oxidative stress can also lead to sustained ion channel-mediated Ca2+ influx and precipitate cytoskeletal disorganization. The complex interplay between GPCR (G-protein coupled receptor) signaling, ion channel activation, and redox injury pathways underscores the need for additional research aimed at identifying targeted therapies to protect podocytes and preserve glomerular function. Earlier detection of albuminuria and podocyte injury through routine noninvasive diagnostics will also be critical in populations at the highest risk for the development of hypertensive kidney disease. In this review, we highlight the established mechanisms of oxidative stress-mediated podocyte damage in proteinuric kidney diseases, with an emphasis on a hypertensive renal injury. We will also consider emerging therapies that have the potential to selectively protect podocytes from redox-related injury.
Collapse
Affiliation(s)
- Daria V. Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Amanda Behr
- Department of Medical Illustration, College of Allied Health Sciences, Augusta University, Augusta, GA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL
- James A. Haley Veterans’ Hospital, Tampa, FL
| | - Gentzon Hall
- Division of Nephrology, Department of Internal Medicine, Duke University School of Medicine, Durham, NC
- Department of Medicine, Division of Nephrology, Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Oleg Palygin
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
3
|
Na DH, Cui S, Fang X, Lee H, Eum SH, Shin YJ, Lim SW, Yang CW, Chung BH. Advancements in Research on Genetic Kidney Diseases Using Human-Induced Pluripotent Stem Cell-Derived Kidney Organoids. Cells 2024; 13:1190. [PMID: 39056771 PMCID: PMC11274677 DOI: 10.3390/cells13141190] [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: 05/14/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Genetic or hereditary kidney disease stands as a pivotal cause of chronic kidney disease (CKD). The proliferation and widespread utilization of DNA testing in clinical settings have notably eased the diagnosis of genetic kidney diseases, which were once elusive but are now increasingly identified in cases previously deemed CKD of unknown etiology. However, despite these diagnostic strides, research into disease pathogenesis and novel drug development faces significant hurdles, chiefly due to the dearth of appropriate animal models and the challenges posed by limited patient cohorts in clinical studies. Conversely, the advent and utilization of human-induced pluripotent stem cells (hiPSCs) offer a promising avenue for genetic kidney disease research. Particularly, the development of hiPSC-derived kidney organoid systems presents a novel platform for investigating various forms of genetic kidney diseases. Moreover, the integration of the CRISPR/Cas9 technique into this system holds immense potential for efficient research on genetic kidney diseases. This review aims to explore the applications of in vitro kidney organoids generated from hiPSCs in the study of diverse genetic kidney diseases. Additionally, it will delve into the limitations of this research platform and outline future perspectives for advancing research in this crucial area.
Collapse
Affiliation(s)
- Do Hyun Na
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sheng Cui
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Xianying Fang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Hanbi Lee
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang Hun Eum
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Incheon 21431, Republic of Korea
| | - Yoo Jin Shin
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Sun Woo Lim
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Chul Woo Yang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Byung Ha Chung
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
4
|
Laffey M, Ashwat E, Lui H, Zhang X, Kaltenmeier C, Packiaraj G, Crane A, Alshamery S, Gunabushanam V, Ganoza A, Dharmayan S, Powers CA, Jonassaint N, Molinari M. Donor-recipient race-ethnicity concordance and patient survival after liver transplantation. HPB (Oxford) 2024; 26:772-781. [PMID: 38523016 DOI: 10.1016/j.hpb.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
INTRODUCTION We assessed the association between patient survival after liver transplantation (LT) and donor-recipient race-ethnicity (R/E) concordance. METHODS The Scientific Registry of Transplant Recipients (SRTR) was retrospectively analyzed using data collected between 2002 and 2019. Only adults without history of prior organ transplant and recipients of LT alone were included. The primary outcome was patient survival. Donors and recipients were categorized into five R/E groups: White/Caucasian, African American/Black, Hispanic/Latino, Asian, and Others. Statistical analyses were performed using Kaplan-Meier survival curves and Cox Proportional Hazards models, adjusting for donor and recipient covariates. RESULTS 85,427 patients were included. Among all the R/E groups, Asian patients had the highest 5-year survival (81.3%; 95% CI = 79.9-82.7), while African American/Black patients had the lowest (71.4%; 95% CI = 70.3-72.6) (P < 0.001). Lower survival rates were observed in recipients who received discordant R/E grafts irrespective of their R/E group. The fully adjusted hazard ratio for death was statistically significant in African American/Black (aHR 1.07-1.18-1.31; P < 0.01) and in White∕Caucasian patients (aHR 1.00-1.04-1.07; P = 0.03) in the presence of donor-recipient R/E discordance. CONCLUSION Disparities in post-LT outcomes might be influenced by biological factors in addition to well-known social determinants of health.
Collapse
Affiliation(s)
- Makenna Laffey
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Eishan Ashwat
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Hao Lui
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Xingyu Zhang
- University of Pittsburgh, School of Health, and Rehabilitation Sciences, Pittsburgh, PA, United States
| | - Christof Kaltenmeier
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Godwin Packiaraj
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Andrew Crane
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Sarmad Alshamery
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Vikraman Gunabushanam
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Armando Ganoza
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Stalin Dharmayan
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Colin A Powers
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States
| | - Naudia Jonassaint
- University of Pittsburgh Medical Center, Department of Medicine, Pittsburgh, PA, United States
| | - Michele Molinari
- University of Pittsburgh Medical Center, Department of Surgery, Pittsburgh, PA, United States.
| |
Collapse
|
5
|
Mitrotti A, Giliberti M, Di Leo V, di Bari I, Pontrelli P, Gesualdo L. Hidden genetics behind glomerular scars: an opportunity to understand the heterogeneity of focal segmental glomerulosclerosis? Pediatr Nephrol 2024; 39:1685-1707. [PMID: 37728640 PMCID: PMC11026212 DOI: 10.1007/s00467-023-06046-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/02/2023] [Accepted: 05/30/2023] [Indexed: 09/21/2023]
Abstract
Focal segmental glomerulosclerosis (FSGS) is a complex disease which describes different kinds of kidney defects, not exclusively linked with podocyte defects. Since nephrin mutation was first described in association with early-onset nephrotic syndrome (NS), many advancements have been made in understanding genetic patterns associated with FSGS. New genetic causes of FSGS have been discovered, displaying unexpected genotypes, and recognizing possible site of damage. Many recent large-scale sequencing analyses on patients affected by idiopathic chronic kidney disease (CKD), kidney failure (KF) of unknown origin, or classified as FSGS, have revealed collagen alpha IV genes, as one of the most frequent sites of pathogenic mutations. Also, recent interest in complex and systemic lysosomal storage diseases, such as Fabry disease, has highlighted GLA mutations as possible causes of FSGS. Tubulointerstitial disease, recently classified by KDIGO based on genetic subtypes, when associated with UMOD variants, may phenotypically gain FSGS features, as well as ciliopathy genes or others, otherwise leading to completely different phenotypes, but found carrying pathogenic variants with associated FSGS phenotype. Thus, glomerulosclerosis may conceal different heterogeneous conditions. When a kidney biopsy is performed, the principal objective is to provide an accurate diagnosis. The broad spectrum of phenotypic expression and genetic complexity is demonstrating that a combined path of management needs to be applied. Genetic investigation should not be reserved only to selected cases, but rather part of medical management, integrating with clinical and renal pathology records. FSGS heterogeneity should be interpreted as an interesting opportunity to discover new pathways of CKD, requiring prompt genotype-phenotype correlation. In this review, we aim to highlight how FSGS represents a peculiar kidney condition, demanding multidisciplinary management, and in which genetic analysis may solve some otherwise unrevealed idiopathic cases. Unfortunately there is not a uniform correlation between specific mutations and FSGS morphological classes, as the same variants may be identified in familial cases or sporadic FSGS/NS or manifest a variable spectrum of the same disease. These non-specific features make diagnosis challenging. The complexity of FSGS genotypes requires new directions. Old morphological classification does not provide much information about the responsible cause of disease and misdiagnoses may expose patients to immunosuppressive therapy side effects, mistaken genetic counseling, and misguided kidney transplant programs.
Collapse
Affiliation(s)
- Adele Mitrotti
- Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy.
| | - Marica Giliberti
- Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Di Leo
- Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Ighli di Bari
- Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Paola Pontrelli
- Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Loreto Gesualdo
- Precision and Regenerative Medicine and Ionian Area, Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
6
|
Dogan M, Watkins C, Ingram H, Moore N, Rucker GM, Gower EG, Eason JD, Bhalla A, Talwar M, Nezakatgoo N, Eymard C, Helmick R, Vanatta J, Bajwa A, Kuscu C, Kuscu C. Unveiling APOL1 haplotypes in a predominantly African-American cohort of kidney transplant patients: a novel classification using probe-independent quantitative real-time PCR. Front Med (Lausanne) 2024; 11:1325128. [PMID: 38660426 PMCID: PMC11039853 DOI: 10.3389/fmed.2024.1325128] [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: 10/20/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Apolipoprotein-L1 (APOL1) is a primate-specific protein component of high-density lipoprotein (HDL). Two variants of APOL1 (G1 and G2), provide resistance to parasitic infections in African Americans but are also implicated in kidney-related diseases and transplant outcomes in recipients. This study aims to identify these risk variants using a novel probe-independent quantitative real-time PCR method in a high African American recipient cohort. Additionally, it aims to develop a new stratification approach based on a haplotype-centric model. Methods Genomic DNA was extracted from recipient PBMCs using SDS lysis buffer and proteinase K. A quantitative PCR assay with modified forward primers and a common reverse primer enabled us to quantitatively identify single nucleotide polymorphisms (SNPs) and the 6-bp deletion. Additionally, we used Sanger sequencing to verify our QPCR findings. Results Our novel probe-independent qPCR effectively distinguished homozygous wild-type, heterozygous SNPs/deletions, and homozygous SNPs/deletions, with at least 4-fold differences. A high prevalence of APOL1 variants was observed (18% two-risk alleles, 34% one-risk allele) in our recipient cohort. Intriguingly, no significant impact of recipient APOL1 variants on transplant outcomes was observed up to 12-month of follow-ups. Ongoing research will encompass more time points and a larger patient cohort, allowing for a comprehensive evaluation of G1/G2 variant subgroups categorized by new haplotype scores, enriching our understanding. Conclusion Our cost-effective and rapid qPCR technique facilitates APOL1 genotyping within hours. Prospective and retrospective studies will enable comparisons with long-term allograft rejection, potentially predicting early/late-stage transplant outcomes based on haplotype evaluation in this diverse group of kidney transplant recipients.
Collapse
Affiliation(s)
- Murat Dogan
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Christine Watkins
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Holly Ingram
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nicholas Moore
- Transplant Research Institute, Memphis, TN, United States
| | - Grace M. Rucker
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | | | | | - Anshul Bhalla
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Manish Talwar
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Nosratollah Nezakatgoo
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Corey Eymard
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Ryan Helmick
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Jason Vanatta
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Methodist Hospital, Memphis, TN, United States
| | - Amandeep Bajwa
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Canan Kuscu
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Cem Kuscu
- Transplant Research Institute, Memphis, TN, United States
- Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
7
|
Gordon EJ, Gacki-Smith J, Gooden MJ, Waite P, Yacat R, Abubakari ZR, Duquette D, Agrawal A, Friedewald J, Savage SK, Cooper M, Gilbert A, Muhammad LN, Wicklund C. Development of a culturally targeted chatbot to inform living kidney donor candidates of African ancestry about APOL1 genetic testing: a mixed methods study. J Community Genet 2024; 15:205-216. [PMID: 38349598 PMCID: PMC11031529 DOI: 10.1007/s12687-024-00698-8] [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: 07/11/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Clinical chatbots are increasingly used to help integrate genetic testing into clinical contexts, but no chatbot exists for Apolipoprotein L1 (APOL1) genetic testing of living kidney donor (LKD) candidates of African ancestry. Our study aimed to culturally adapt and assess perceptions of the Gia® chatbot to help integrate APOL1 testing into LKD evaluation. Ten focus groups and post-focus group surveys were conducted with 54 LKDs, community members, and kidney transplant recipients of African ancestry. Data were analyzed through thematic analysis and descriptive statistics. Key themes about making Gia culturally targeted included ensuring: (1) transparency by providing Black LKDs' testimonials, explaining patient privacy and confidentiality protections, and explaining how genetic testing can help LKD evaluation; (2) content is informative by educating Black LKDs about APOL1 testing instead of aiming to convince them to undergo testing, presenting statistics, and describing how genetic discrimination is legally prevented; and (3) content avoids stigma about living donation in the Black community. Most agreed Gia was neutral and unbiased (82%), trustworthy (82%), and words, phrases, and expressions were familiar to the intended audience (85%). Our culturally adapted APOL1 Gia chatbot was well regarded. Future research should assess how this chatbot could supplement provider discussion prior to genetic testing to scale APOL1 counseling and testing for LKD candidate clinical evaluation.
Collapse
Affiliation(s)
- Elisa J Gordon
- Department of Surgery, Center for Biomedical Ethics and Society, Vanderbilt University Medical Center, 1161 21St Avenue South, D-4314 Medical Center North Nashville, Nashville, TN, 37232-2730, USA.
| | - Jessica Gacki-Smith
- Center for Health Services and Outcomes Research, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew J Gooden
- Center for Health Services and Outcomes Research, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Preeya Waite
- Center for Health Services and Outcomes Research, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rochell Yacat
- Medstar Georgetown Transplant Institute, Georgetown University Hospital, Washington, DC, USA
| | - Zenab R Abubakari
- Medstar Georgetown Transplant Institute, Georgetown University Hospital, Washington, DC, USA
| | - Debra Duquette
- Medicine, Cardiology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Akansha Agrawal
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - John Friedewald
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Matthew Cooper
- Froedtert Hospital Center for Advanced Care, Froedtert Memorial Lutheran Hospital Children's Hospital of Wisconsin Medical College of Wisconsin, Milwaukee, WI, USA
- Children's Hospital of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin, Milwaukee, WI, USA
- Froedtert Hospital Center for Advanced Care, Milwaukee, WI, USA
| | - Alexander Gilbert
- Medstar Georgetown Transplant Institute, Georgetown University Hospital, Washington, DC, USA
| | - Lutfiyya N Muhammad
- Department of Preventive Medicine, Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Catherine Wicklund
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
8
|
Lo R, Narasaki Y, Lei S, Rhee CM. Management of traditional risk factors for the development and progression of chronic kidney disease. Clin Kidney J 2023; 16:1737-1750. [PMID: 37915906 PMCID: PMC10616454 DOI: 10.1093/ckj/sfad101] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Indexed: 11/03/2023] Open
Abstract
Chronic kidney disease (CKD) and its downstream complications (i.e. cardiovascular) are a major source of morbidity worldwide. Additionally, deaths due to CKD or CKD-attributable cardiovascular disease account for a sizeable proportion of global mortality. However, the advent of new pharmacotherapies, diagnostic tools, and global initiatives are directing greater attention to kidney health in the public health agenda, including the implementation of effective strategies that (i) prevent kidney disease, (ii) provide early CKD detection, and (iii) ameliorate CKD progression and its related complications. In this Review, we discuss major risk factors for incident CKD and CKD progression categorized across cardiovascular (i.e. hypertension, dyslipidemia, cardiorenal syndrome), endocrine (i.e. diabetes mellitus, hypothyroidism, testosterone), lifestyle (i.e. obesity, dietary factors, smoking), and genetic/environmental (i.e. CKDu/Mesoamerican nephropathy, APOL1, herbal nephropathy) domains, as well as scope, mechanistic underpinnings, and management.
Collapse
Affiliation(s)
- Robin Lo
- Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine, Orange, CA, USA
| | - Yoko Narasaki
- Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine, Orange, CA, USA
- Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA, USA
| | - Sean Lei
- Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine, Orange, CA, USA
| | - Connie M Rhee
- Harold Simmons Center for Chronic Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine, Orange, CA, USA
- Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA, USA
| |
Collapse
|
9
|
Dogan M, Watkins C, Ingram H, Moore N, Rucker GM, Gower EG, Eason JD, Bhalla A, Talwar M, Nezakatgoo N, Eymard C, Helmick R, Vanatta J, Bajwa A, Kuscu C, Kuscu C. Unveiling APOL1 Haplotypes: A Novel Classification Through Probe-Independent Quantitative Real-Time PCR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562539. [PMID: 37905084 PMCID: PMC10614821 DOI: 10.1101/2023.10.16.562539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Introduction Apolipoprotein-L1 (APOL1) is a primate-specific protein component of high- density lipoprotein (HDL). Two variants of APOL1 (G1 and G2), provide resistance to parasitic infections in African Americans but are also implicated in kidney-related diseases and transplant outcomes in recipients. This study aims to identify these risk variants using a novel probe- independent quantitative real-time PCR method in a high African American recipient cohort. Additionally, it aims to develop a new stratification approach based on haplotype-centric model. Methods Genomic DNA was extracted from recipient PBMCs using SDS lysis buffer and proteinase K. Quantitative PCR assay with modified forward primers and a common reverse primer enabled us to identify single nucleotide polymorphisms (SNPs) and the 6-bp deletion quantitatively. Additionally, we used sanger sequencing to verify our QPCR findings. Results Our novel probe-independent qPCR effectively distinguished homozygous wild-type, heterozygous SNPs/deletion, and homozygous SNPs/deletion, with at least 4-fold differences. High prevalence of APOL1 variants was observed (18% two-risk alleles, 34% one-risk allele) in our recipient cohort. Intriguingly, up to 12-month follow-up revealed no significant impact of recipient APOL1 variants on transplant outcomes. Ongoing research will encompass more time points and a larger patient cohort, allowing a comprehensive evaluation of G1/G2 variant subgroups categorized by new haplotype scores, enriching our understanding. Conclusions Our cost-effective and rapid qPCR technique facilitates APOL1 genotyping within hours. Prospective and retrospective studies will enable comparisons with long-term allograft rejection, potentially predicting early/late-stage transplant outcomes based on haplotype evaluation in this diverse group of kidney transplant recipients.
Collapse
|
10
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
11
|
Giudicelli GC, De Souza CMB, Veronese FV, Pereira LV, Hünemeier T, Vianna FSL. Precision medicine implementation challenges for APOL1 testing in chronic kidney disease in admixed populations. Front Genet 2022; 13:1016341. [PMID: 36588788 PMCID: PMC9797503 DOI: 10.3389/fgene.2022.1016341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Chronic Kidney Disease (CKD) is a public health problem that presents genetic and environmental risk factors. Two alleles in the Apolipoprotein L1 (APOL1) gene were associated with chronic kidney disease; these alleles are common in individuals of African ancestry but rare in European descendants. Genomic studies on Afro-Americans have indicated a higher prevalence and severity of chronic kidney disease in people of African ancestry when compared to other ethnic groups. However, estimates in low- and middle-income countries are still limited. Precision medicine approaches could improve clinical outcomes in carriers of risk alleles in the Apolipoprotein L1 gene through early diagnosis and specific therapies. Nevertheless, to enhance the definition of studies on these variants, it would be necessary to include individuals with different ancestry profiles in the sample, such as Latinos, African Americans, and Indigenous peoples. There is evidence that measuring genetic ancestry improves clinical care for admixed people. For chronic kidney disease, this knowledge could help establish public health strategies for monitoring patients and understanding the impact of the Apolipoprotein L1 genetic variants in admixed populations. Therefore, researchers need to develop resources, methodologies, and incentives for vulnerable and disadvantaged communities, to develop and implement precision medicine strategies and contribute to consolidating diversity in science and precision medicine in clinical practice.
Collapse
Affiliation(s)
- Giovanna Câmara Giudicelli
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Porto Alegre, RS, Brazil
| | - Celia Mariana Barbosa De Souza
- Departamento de Nefrologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Francisco Veríssimo Veronese
- Departamento de Nefrologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-graduação em Medicina: Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lygia V. Pereira
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Tábita Hünemeier
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
- Institut de Biologia Evolutiva, CSIC/Universitat Pompeu Fabra, Barcelona, Spain
| | - Fernanda Sales Luiz Vianna
- Laboratório de Medicina Genômica, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência e Tecnologia de Genética Médica Populacional, Porto Alegre, RS, Brazil
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Medicina Personalizada Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
12
|
Ilori T, Watanabe A, Ng KH, Solarin A, Sinha A, Gbadegesin R. Genetics of Chronic Kidney Disease in Low-Resource Settings. Semin Nephrol 2022; 42:151314. [PMID: 36801667 PMCID: PMC10272019 DOI: 10.1016/j.semnephrol.2023.151314] [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] [Indexed: 02/19/2023]
Abstract
Advances in kidney genomics in the past 20 years has opened the door for more precise diagnosis of kidney disease and identification of new and specific therapeutic agents. Despite these advances, an imbalance exists between low-resource and affluent regions of the world. Individuals of European ancestry from the United States, United Kingdom, and Iceland account for 16% of the world's population, but represent more than 80% of all genome-wide association studies. South Asia, Southeast Asia, Latin America, and Africa together account for 57% of the world population but less than 5% of genome-wide association studies. Implications of this difference include limitations in new variant discovery, inaccurate interpretation of the effect of genetic variants in non-European populations, and unequal access to genomic testing and novel therapies in resource-poor regions. It also further introduces ethical, legal, and social pitfalls, and ultimately may propagate global health inequities. Ongoing efforts to reduce the imbalance in low-resource regions include funding and capacity building, population-based genome sequencing, population-based genome registries, and genetic research networks. More funding, training, and capacity building for infrastructure and expertise is needed in resource-poor regions. Focusing on this will ensure multiple-fold returns on investments in genomic research and technology.
Collapse
Affiliation(s)
- Titilayo Ilori
- Division of Nephrology, Boston University School of Medicine, Boston, MA
| | - Andreia Watanabe
- Division of Molecular Medicine, Department of Pediatrics, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Kar-Hui Ng
- Department of Pediatrics, Yong Loo Lin School of Medicine, Singapore
| | - Adaobi Solarin
- Department of Pediatrics and Child Health, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria
| | - Aditi Sinha
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Rasheed Gbadegesin
- Division of Nephrology, Department of Pediatrics, Duke University School of Medicine, Durham, NC.
| |
Collapse
|
13
|
Wu M, Chen Y, Chiu I, Wu M. Genetic Insight into Primary Glomerulonephritis. Nephrology (Carlton) 2022; 27:649-657. [DOI: 10.1111/nep.14074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/18/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Mei‐Yi Wu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health National Taiwan University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
| | - Ying‐Chun Chen
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
| | - I‐Jen Chiu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
| | - Mai‐Szu Wu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine Taipei Medical University Taipei Taiwan
| |
Collapse
|
14
|
Genetics in chronic kidney disease: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int 2022; 101:1126-1141. [PMID: 35460632 PMCID: PMC9922534 DOI: 10.1016/j.kint.2022.03.019] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/16/2022] [Accepted: 03/29/2022] [Indexed: 01/19/2023]
Abstract
Numerous genes for monogenic kidney diseases with classical patterns of inheritance, as well as genes for complex kidney diseases that manifest in combination with environmental factors, have been discovered. Genetic findings are increasingly used to inform clinical management of nephropathies, and have led to improved diagnostics, disease surveillance, choice of therapy, and family counseling. All of these steps rely on accurate interpretation of genetic data, which can be outpaced by current rates of data collection. In March of 2021, Kidney Diseases: Improving Global Outcomes (KDIGO) held a Controversies Conference on "Genetics in Chronic Kidney Disease (CKD)" to review the current state of understanding of monogenic and complex (polygenic) kidney diseases, processes for applying genetic findings in clinical medicine, and use of genomics for defining and stratifying CKD. Given the important contribution of genetic variants to CKD, practitioners with CKD patients are advised to "think genetic," which specifically involves obtaining a family history, collecting detailed information on age of CKD onset, performing clinical examination for extrarenal symptoms, and considering genetic testing. To improve the use of genetics in nephrology, meeting participants advised developing an advanced training or subspecialty track for nephrologists, crafting guidelines for testing and treatment, and educating patients, students, and practitioners. Key areas of future research, including clinical interpretation of genome variation, electronic phenotyping, global representation, kidney-specific molecular data, polygenic scores, translational epidemiology, and open data resources, were also identified.
Collapse
|
15
|
Yang YW, Poudel B, Frederick J, Dhillon P, Shrestha R, Ma Z, Wu J, Okamoto K, Kopp JB, Booten SL, Gattis D, Watt AT, Palmer M, Aghajan M, Susztak K. Antisense oligonucleotides ameliorate kidney dysfunction in podocyte specific APOL1 risk variant mice. Mol Ther 2022; 30:2491-2504. [PMID: 35450819 DOI: 10.1016/j.ymthe.2022.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 03/23/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022] Open
Abstract
Coding variants (named G1 and G2) in Apolipoprotein L1 (APOL1) can explain the most excess risk of kidney disease observed in African Americans. It has been proposed that risk variant APOL1 dose, such as increased risk variant APOL1 level serves as a trigger (second hit) for disease development. The goal of this study was to determine whether lowering risk variant APOL1 levels protects from disease development in podocyte specific transgenic mouse disease model. We administered antisense oligonucleotides (ASO) targeting APOL1 to podocyte specific G2APOL1 mice and observed efficient reduction of APOL1 levels. APOL1 ASO1, which more efficiently lowered APOL1 transcript levels, protected mice from albuminuria, glomerulosclerosis, tubulointerstitial fibrosis, and renal failure. The administration of APOL1 ASO1 was effective even for established disease in the NEFTA-rtTA/TRE-G2APOL1 (NEFTA/G2APOL1) mice. We observed a strong correlation between APOL1 transcript level and disease severity. We concluded that an APOL1 ASO1 may be an effective therapeutic approach for APOL1-associated glomerular disease.
Collapse
Affiliation(s)
- Ya-Wen Yang
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Bibek Poudel
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Julia Frederick
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Poonam Dhillon
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Rojesh Shrestha
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ziyuan Ma
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Junnan Wu
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Koji Okamoto
- Kidney Disease Section, NIDDK, NIH, Bethesda, MD, USA
| | | | | | | | | | - Matthew Palmer
- Department of Pathology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Katalin Susztak
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Hung AM, Shah SC, Bick AG, Yu Z, Chen HC, Hunt CM, Wendt F, Wilson O, Greevy RA, Chung CP, Suzuki A, Ho YL, Akwo E, Polimanti R, Zhou J, Reaven P, Tsao PS, Gaziano JM, Huffman JE, Joseph J, Luoh SW, Iyengar S, Chang KM, Casas JP, Matheny ME, O’Donnell CJ, Cho K, Tao R, Susztak K, Robinson-Cohen C, Tuteja S, Siew ED. APOL1 Risk Variants, Acute Kidney Injury, and Death in Participants With African Ancestry Hospitalized With COVID-19 From the Million Veteran Program. JAMA Intern Med 2022; 182:386-395. [PMID: 35089317 PMCID: PMC8980930 DOI: 10.1001/jamainternmed.2021.8538] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/25/2021] [Indexed: 01/30/2023]
Abstract
IMPORTANCE Coronavirus disease 2019 (COVID-19) confers significant risk of acute kidney injury (AKI). Patients with COVID-19 with AKI have high mortality rates. OBJECTIVE Individuals with African ancestry with 2 copies of apolipoprotein L1 (APOL1) variants G1 or G2 (high-risk group) have significantly increased rates of kidney disease. We tested the hypothesis that the APOL1 high-risk group is associated with a higher-risk of COVID-19-associated AKI and death. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study included 990 participants with African ancestry enrolled in the Million Veteran Program who were hospitalized with COVID-19 between March 2020 and January 2021 with available genetic information. EXPOSURES The primary exposure was having 2 APOL1 risk variants (RV) (APOL1 high-risk group), compared with having 1 or 0 risk variants (APOL1 low-risk group). MAIN OUTCOMES AND MEASURES The primary outcome was AKI. The secondary outcomes were stages of AKI severity and death. Multivariable logistic regression analyses adjusted for preexisting comorbidities, medications, and inpatient AKI risk factors; 10 principal components of ancestry were performed to study these associations. We performed a subgroup analysis in individuals with normal kidney function prior to hospitalization (estimated glomerular filtration rate ≥60 mL/min/1.73 m2). RESULTS Of the 990 participants with African ancestry, 905 (91.4%) were male with a median (IQR) age of 68 (60-73) years. Overall, 392 (39.6%) patients developed AKI, 141 (14%) developed stages 2 or 3 AKI, 28 (3%) required dialysis, and 122 (12.3%) died. One hundred twenty-five (12.6%) of the participants were in the APOL1 high-risk group. Patients categorized as APOL1 high-risk group had significantly higher odds of AKI (adjusted odds ratio [OR], 1.95; 95% CI, 1.27-3.02; P = .002), higher AKI severity stages (OR, 2.03; 95% CI, 1.37-2.99; P < .001), and death (OR, 2.15; 95% CI, 1.22-3.72; P = .007). The association with AKI persisted in the subgroup with normal kidney function (OR, 1.93; 95% CI, 1.15-3.26; P = .01). Data analysis was conducted between February 2021 and April 2021. CONCLUSIONS AND RELEVANCE In this cohort study of veterans with African ancestry hospitalized with COVID-19 infection, APOL1 kidney risk variants were associated with higher odds of AKI, AKI severity, and death, even among individuals with prior normal kidney function.
Collapse
Affiliation(s)
- Adriana M. Hung
- Tennessee Valley Healthcare System, Nashville Campus, Nashville
- Division of Nephrology & Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shailja C. Shah
- GI Section, VA San Diego Healthcare System, San Diego, California
- Division of Gastroenterology, University of California, San Diego, San Diego
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Zhihong Yu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christine M. Hunt
- Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina
- VA Cooperative Studies Program Epidemiology Center, Durham VA Health Care System, Durham, North Carolina
| | - Frank Wendt
- Department of Psychiatry, Yale University School of Medicine, West Haven, Connecticut
- VA CT Healthcare Center, West Haven, Connecticut
| | - Otis Wilson
- Division of Nephrology & Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert A. Greevy
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cecilia P. Chung
- Division of Rheumatology and Division of Clinical Pharmacology, Vanderbilt University Medical Center, Rheumatology Section, Veterans Affairs, Nashville, Tennessee
| | - Ayako Suzuki
- Division of Gastroenterology, Duke University Medical Center, Durham, North Carolina
- VA Cooperative Studies Program Epidemiology Center, Durham VA Health Care System, Durham, North Carolina
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston
| | - Elvis Akwo
- Division of Nephrology & Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, West Haven, Connecticut
- VA CT Healthcare Center, West Haven, Connecticut
| | - Jin Zhou
- Department of Epidemiology and Biostatistics, University of Arizona, Phoenix
- Phoenix VA Health Care System, Phoenix, Arizona
| | - Peter Reaven
- Phoenix VA Health Care System, Phoenix, Arizona
- Division of Endocrinology, Department of Medicine, University of Arizona, Phoenix
| | - Philip S. Tsao
- Epidemiology Research and Information Center (ERIC), VA Palo Alto Health Care System, Palo Alto, California
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California
| | - J. Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston
- Division of Aging, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Jennifer E. Huffman
- Center for Population Genomics, Massachusetts Veterans Epidemiology Research & Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts
| | - Jacob Joseph
- Cardiology Section, Veterans Affairs Boston, Boston, Massachusetts
- Division of Cardiovascular Medicine, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Shiuh-Wen Luoh
- VA Portland Health Care System, Portland, Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Sudha Iyengar
- Department of Population and Quantitative Health Sciences, Case Western Reserve University and Louis Stoke, Cleveland VA, Cleveland, Ohio
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio
| | - Kyong-Mi Chang
- The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
| | - Juan P. Casas
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston
- Department of Medicine, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Michael E. Matheny
- Departments of Biomedical Informatics, Biostatistics, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- GREEC, TVHS VA, Nashville, Tennessee
| | - Christopher J. O’Donnell
- Cardiology, VA Boston Healthcare System, Boston, Massachusetts
- Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Novartis
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston
- Department of Medicine, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Cassianne Robinson-Cohen
- Division of Nephrology & Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sony Tuteja
- The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward D. Siew
- Division of Nephrology & Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Healthcare System, Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|
17
|
Piras D, Lepori N, Cabiddu G, Pani A. How Genetics Can Improve Clinical Practice in Chronic Kidney Disease: From Bench to Bedside. J Pers Med 2022; 12:jpm12020193. [PMID: 35207681 PMCID: PMC8875178 DOI: 10.3390/jpm12020193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Chronic kidney disease (CKD) is considered a major global health problem with high socio-economic costs: the risk of CKD in individuals with an affected first degree relative has been found to be three times higher than in the general population. Genetic factors are known to be involved in CKD pathogenesis, both due to the possible presence of monogenic pathologies as causes of CKD, and to the role of numerous gene variants in determining susceptibility to the development of CKD. The genetic study of CKD patients can represent a useful tool in the hands of the clinician; not only in the diagnostic and prognostic field, but potentially also in guiding therapeutic choices and in designing clinical trials. In this review we discuss the various aspects of the role of genetic analysis on clinical management of patients with CKD with a focus on clinical applications. Several topics are discussed in an effort to provide useful information for daily clinical practice: definition of susceptibility to the development of CKD, identification of unrecognized monogenic diseases, reclassification of the etiological diagnosis, role of pharmacogenetics.
Collapse
Affiliation(s)
- Doloretta Piras
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
- Correspondence:
| | - Nicola Lepori
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
| | - Gianfranca Cabiddu
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, 09134 Cagliari, Italy
| | - Antonello Pani
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, 09134 Cagliari, Italy
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerce (CNR), 09042 Monserrato, Italy
| |
Collapse
|
18
|
Comparative Analysis of the APOL1 Variants in the Genetic Landscape of Renal Carcinoma Cells. Cancers (Basel) 2022; 14:cancers14030733. [PMID: 35159001 PMCID: PMC8833631 DOI: 10.3390/cancers14030733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Renal cell carcinoma (RCC) occurs at higher frequency in individuals of African ancestry, with well-recorded documentation in this community. This is most prominent in the context of chronic kidney disease. In turn, many forms of progressive chronic kidney disease are more common in populations of Sub-Saharan African ancestry. This disparity has been attributed to well-defined allelic variants and has risen in the parental populations to high frequency under evolutionary pressure. Mechanisms of increased kidney disease risk and cell injury, causally associated with these APOL1 gene variants, have been extensively studied. Most studies have compared the effects of ectopic overexpression of the parental non-risk APOL1 with the mutated risk variants in cellular and organismal platforms. In the current study, we have used CRISPR/Cas9 genetic engineering to knock out or modify the sequence of endogenous APOL1 in RCC to mimic and examine the effects of these naturally occurring kidney disease risk allelic variants. Remarkably, these modifications to endogenous APOL1 genes in RCC resulted in a set of prominent effects on mitochondrial integrity and metabolic pathways and disrupted tumorigenesis. These findings both clarify pathways of cell injury of APOL1 risk variants in cells of kidney origin and motivate further studies to examine the potential central role of APOL1 in the pathogenesis of renal cell carcinoma and its relation to chronic kidney disease in genotypically at-risk African ancestry individuals. Abstract Although the relative risk of renal cell carcinoma associated with chronic kidney injury is particularly high among sub-Saharan African ancestry populations, it is unclear yet whether the APOL1 gene risk variants (RV) for kidney disease additionally elevate this risk. APOL1 G1 and G2 RV contribute to increased risk for kidney disease in black populations, although the disease mechanism has still not been fully deciphered. While high expression levels of all three APOL1 allelic variants, G0 (the wild type allele), G1, and G2 are injurious to normal human cells, renal carcinoma cells (RCC) naturally tolerate inherent high expression levels of APOL1. We utilized CRISPR/Cas9 gene editing to generate isogenic RCC clones expressing APOL1 G1 or G2 risk variants on a similar genetic background, thus enabling a reliable comparison between the phenotypes elicited in RCC by each of the APOL1 variants. Here, we demonstrate that knocking in the G1 or G2 APOL1 alleles, or complete elimination of APOL1 expression, has major effects on proliferation capacity, mitochondrial morphology, cell metabolism, autophagy levels, and the tumorigenic potential of RCC cells. The most striking effect of the APOL1 RV effect was demonstrated in vivo by the complete abolishment of tumor growth in immunodeficient mice. Our findings suggest that, in contrast to the WT APOL1 variant, APOL1 RV are toxic for RCC cells and may act to suppress cancer cell growth. We conclude that the inherent expression of non-risk APOL1 G0 is required for RCC tumorigenicity. RCC cancer cells can hardly tolerate increased APOL1 risk variants expression levels as opposed to APOL1 G0.
Collapse
|
19
|
Müller D, Schmitz J, Fischer K, Granado D, Groh AC, Krausel V, Lüttgenau SM, Amelung TM, Pavenstädt H, Weide T. Evolution of Renal-Disease Factor APOL1 Results in Cis and Trans Orientations at the Endoplasmic Reticulum That Both Show Cytotoxic Effects. Mol Biol Evol 2021; 38:4962-4976. [PMID: 34323996 PMCID: PMC8557400 DOI: 10.1093/molbev/msab220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The recent and exclusively in humans and a few other higher primates expressed APOL1 (apolipoprotein L1) gene is linked to African human trypanosomiasis (also known as African sleeping sickness) as well as to different forms of kidney diseases. Whereas APOL1's role as a trypanolytic factor is well established, pathobiological mechanisms explaining its cytotoxicity in renal cells remain unclear. In this study, we compared the APOL family members using a combination of evolutionary studies and cell biological experiments to detect unique features causal for APOL1 nephrotoxic effects. We investigated available primate and mouse genome and transcriptome data to apply comparative phylogenetic and maximum likelihood selection analyses. We suggest that the APOL gene family evolved early in vertebrates and initial splitting occurred in ancestral mammals. Diversification and differentiation of functional domains continued in primates, including developing the two members APOL1 and APOL2. Their close relationship could be diagnosed by sequence similarity and a shared ancestral insertion of an AluY transposable element. Live-cell imaging analyses showed that both expressed proteins show a strong preference to localize at the endoplasmic reticulum (ER). However, glycosylation and secretion assays revealed that-unlike APOL2-APOL1 membrane insertion or association occurs in different orientations at the ER, with the disease-associated mutants facing either the luminal (cis) or cytoplasmic (trans) side of the ER. The various pools of APOL1 at the ER offer a novel perspective in explaining the broad spectrum of its observed toxic effects.
Collapse
Affiliation(s)
- Daria Müller
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Jürgen Schmitz
- Institute of Experimental Pathology, ZMBE, University of Münster, Münster, Germany
| | - Katharina Fischer
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Daniel Granado
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Ann-Christin Groh
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Vanessa Krausel
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Simona Mareike Lüttgenau
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Till Maximilian Amelung
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Hermann Pavenstädt
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| | - Thomas Weide
- Internal Medicine D (MedD), Molecular Nephrology, University Hospital of Münster (UKM), Münster, Germany
| |
Collapse
|
20
|
Nutrition Intervention for Reduction of Cardiovascular Risk in African Americans Using the 2019 American College of Cardiology/American Heart Association Primary Prevention Guidelines. Nutrients 2021; 13:nu13103422. [PMID: 34684423 PMCID: PMC8537862 DOI: 10.3390/nu13103422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The 2019 American College of Cardiology/American Heart Association (ACC/AHA) Prevention Guidelines emphasize reduction in dietary sodium, cholesterol, refined carbohydrates, saturated fat and sweetened beverages. We hypothesized that implementing this dietary pattern could reduce cardiovascular risk in a cohort of volunteers in an urban African American (AA) community church, during a 5-week ACC/AHA-styled nutrition intervention, assessed by measuring risk markers and adherence, called HEART-LENS (Helping Everyone Assess Risk Today Lenten Nutrition Study). METHODS The study population consisted of 53 volunteers who committed to eat only home-delivered non-dairy vegetarian meals (average daily calories 1155, sodium 1285 mg, cholesterol 0 mg; 58% carbohydrate, 17% protein, 25% fat). Body mass index (BMI) and fasting serum markers of cardiometabolic and risk factors were measured, with collection of any dietary deviation. RESULTS Of 53 volunteers, 44 (mean age 60.2 years, 37 women) completed the trial (88%); 1 was intolerant of the meals, 1 completed both blood draws but did not eat delivered food, and 7 did not return for the tests. Adherence to the diet was reported at 93% in the remaining 44. Cardiometabolic risk factors improved significantly, highlighted by a marked reduction in serum insulin (-43%, p = 0.000), hemoglobin A1c (6.2% to 6.0%, p = 0.000), weight and BMI (-10.2 lbs, 33 to 31 kg/m2, p = 0.000), but with small reductions of fasting glucose (-6%, p = 0.405) and triglyceride levels (-4%, p = 0.408). Additionally, improved were trimethylamine-N-oxide (5.1 to 2.9 µmol/L, -43%, p = 0.001), small dense low-density lipoprotein cholesterol (LDL) (24.2 to 19.1 mg/dL, -21%, p = 0.000), LDL (121 to 104 mg/dL, -14%, p = 0.000), total cholesterol (TC) (190 to 168 mg/dL, -12%, p = 0.000), and lipoprotein (a) (LP(a)) (56 to 51 mg/dL, -11%, p = 0.000); high sensitivity C-reactive protein (hs-CRP) was widely variable but reduced by 16% (2.5 to 2.1 ng/mL, p = NS) in 40 subjects without inflammatory conditions. Soluble urokinase plasminogen activator (suPAR) levels were not significantly changed. The ACC/AHA pooled cohort atherosclerotic cardiovascular disease (ASCVD) risk scores were calculated for 41 and 36 volunteers, respectively, as the ASCVD risk could not be calculated for 3 subjects with low lipid fractions at baseline and 8 subjects after intervention (p = 0.184). In the remaining subjects, the mean 10-year risk was reduced from 10.8 to 8.7%, a 19.4% decrease (p = 0.006), primarily due to a 14% decrease in low-density lipoprotein cholesterol and a 10 mm Hg (6%) reduction in systolic blood pressure. CONCLUSIONS In this prospective 5-week non-dairy vegetarian nutrition intervention with good adherence consistent with the 2019 ACC/AHA Guidelines in an at-risk AA population, markers of cardiovascular risk, cardiometabolism, and body weight were significantly reduced, including obesity, low-density lipoprotein cholesterol (LDLc) density, LP(a), inflammation, and ingestion of substrates mediating production of trimethylamine-N-oxide (TMAO). Albeit reduced, hs-CRP and suPAR, were not lowered consistently. This induced a significant decrease in the 10-year ASCVD risk in this AA cohort. If widely adopted, this could dramatically reduce and possibly eradicate, the racial disparity in ASCVD events and mortality, if 19% of the 21% increase is eliminated by this lifestyle change.
Collapse
|
21
|
Abstract
Rates of many types of severe kidney disease are much higher in Black individuals than most other ethnic groups. Much of this disparity can now be attributed to genetic variants in the apoL1 (APOL1) gene found only in individuals with recent African ancestry. These variants greatly increase rates of hypertension-associated ESKD, FSGS, HIV-associated nephropathy, and other forms of nondiabetic kidney disease. We discuss the population genetics of APOL1 risk variants and the clinical spectrum of APOL1 nephropathy. We then consider clinical issues that arise for the practicing nephrologist caring for the patient who may have APOL1 kidney disease.
Collapse
Affiliation(s)
- David J Friedman
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
22
|
Affiliation(s)
- Linda T Hiraki
- Pediatrics and Epidemiology, Dalla Lana School of Public Health, University of Toronto, and Clinician Scientist, Division of Rheumatology, and Scientist, Child Health Evaluative Sciences and Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
23
|
Wang DP, Yu ZX, He ZC, Liao JF, Shen XB, Zhu PL, Chen WN, Lin X, Xu SH. Apolipoprotein L1 is transcriptionally regulated by SP1, IRF1 and IRF2 in hepatoma cells. FEBS Lett 2020; 594:3108-3121. [PMID: 32671843 DOI: 10.1002/1873-3468.13887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/21/2020] [Accepted: 07/05/2020] [Indexed: 11/07/2022]
Abstract
Apolipoprotein L1 (APOL1) participates in lipid metabolism. Here, we investigate the mechanisms regulating APOL1 gene expression in hepatoma cells. We demonstrate that the -80-nt to +31-nt region of the APOL1 promoter, which contains one SP transcription factor binding GT box and an interferon regulatory factor (IRF) binding ISRE element, maintains the maximum activity. Mutation of the GT box and ISRE element dramatically reduces APOL1 promoter activity. EMSA and chromatin immunoprecipitation assay reveal that the transcription factors Sp1, IRF1 and IRF2 could interact with their cognate binding sites on the APOL1 promoter. Overexpression of Sp1, IRF1 and IRF2 increases promoter activity, leading to increased APOL1 mRNA and protein levels, while knockdown of Sp1, IRF1 and IRF2 has the opposite effects. These results demonstrate that the APOL1 gene could be regulated by Sp1, IRF1 and IRF2 in hepatoma cells.
Collapse
Affiliation(s)
- De-Ping Wang
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Endocrinology and Metabolism, Hongqi Hospital of MuDanJiang Medical College, Mudanjiang, China
| | - Zhao-Xi Yu
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Zong-Cun He
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jin-Fu Liao
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xue-Bin Shen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, China
| | - Peng-Li Zhu
- Department of Medical Intensive Care Unit, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wan-Nan Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shang-Hua Xu
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping, China
| |
Collapse
|
24
|
Hassan MO, Duarte R, Dickens C, Dix-Peek T, Naidoo S, Vachiat A, Grinter S, Manga P, Naicker S. APOL1 Genetic Variants Are Associated with Serum-Oxidized Low-Density Lipoprotein Levels and Subclinical Atherosclerosis in South African CKD Patients. Nephron Clin Pract 2020; 144:331-340. [PMID: 32526749 DOI: 10.1159/000507860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/13/2020] [Indexed: 02/15/2024] Open
Abstract
INTRODUCTION Apolipoprotein L1 (APOL1) plays an important role in cholesterol metabolism and attenuation of low-density lipoprotein (LDL) oxidation. While protecting against Trypanosoma brucei rhodesiense infection, APOL1 risk alleles confer greater risk for CKD and cardiovascular disease among patients of African descent. OBJECTIVES We investigated whether APOL1 risk variants are associated with atherosclerosis and oxidized LDL (OxLDL) levels among black South African CKD patients. METHODS A cross-sectional study of 120 adult CKD patients and 40 controls was undertaken. DNA samples of participants were genotyped for APOL1 G1 and G2 variants. High-sensitivity C-reactive protein, serum lipids, and OxLDL levels were measured, and carotid doppler ultrasonography was performed on all participants. RESULTS APOL1 alleles rs73885319, rs60910145, and rs71785313 had minor allele frequencies of 9.2, 8.8, and 17.5%, respectively, in the patients, and 8.8, 8.8, and 13.8%, respectively, in the controls. Of the 9 patients with 2 APOL1 risk alleles, 77.8% were compound G1/G2 heterozygotes and 22.2% were G2 homozygotes. Carriers of at least 1 APOL1 risk allele had a 3-fold increased risk of subclinical atherosclerosis (odds ratio 3.19; 95% confidence interval: 1.64-6.19; p = 0.01) compared to individuals with no risk alleles. Patients with 1 or 2 APOL1 risk alleles showed a significant increase in OxLDL levels when compared with those without the APOL1 risk allele. CONCLUSION These findings suggest an increased risk for atherosclerosis in carriers of a single APOL1 risk variant, and the presence of APOL1 risk variants was associated with increased serum OxLDL levels in black South African CKD patients.
Collapse
Affiliation(s)
- Muzamil Olamide Hassan
- Divisions of Nephrology, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa,
| | - Raquel Duarte
- Internal Medicine Research Laboratory, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline Dickens
- Internal Medicine Research Laboratory, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Therese Dix-Peek
- Internal Medicine Research Laboratory, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Sagren Naidoo
- Divisions of Nephrology, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Ahmed Vachiat
- Division of Cardiology, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Sacha Grinter
- Division of Cardiology, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Pravin Manga
- Division of Cardiology, Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Saraladevi Naicker
- Department of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
25
|
Abstract
Apolipoprotein L1 (APOL1) is a protein encoded by the APOL1 gene, found only in humans and several primates. Two variants encoding two different isoforms exist for APOL1, namely G1 and G2. These variants confer increased protection against trypanosome infection, and subsequent African sleeping sickness, and also increase the likelihood of renal disease in individuals of African ancestry. APOL1 mutations are associated with increased risk of chronic kidney disease, inflammation, and exacerbation of systemic lupus erythematosus-associated renal dysfunction. This review serves to outline the structure and function of APOL1, as well as its role in several disease outcomes.
Collapse
Affiliation(s)
- Shanel Raghubeer
- Biomedical Sciences, Cape Peninsula University of Technology-Bellville Campus, Cape Town, Western Cape, South Africa
| | - Tahir S Pillay
- Department of Chemical Pathology, University of Pretoria Faculty of Health Sciences, Pretoria, Gauteng, South Africa.,Division of Chemical Pathology, University of Cape Town, Rondebosch, Western Cape, South Africa
| | - Tandi Edith Matsha
- Biomedical Sciences, Cape Peninsula University of Technology-Bellville Campus, Cape Town, Western Cape, South Africa
| |
Collapse
|
26
|
Abstract
Chronic kidney disease increasingly is being recognized as an important global public health problem. Interindividual susceptibility to kidney disease is high and likely is dependent on risk modulation through genetics, fetal and early childhood development, environmental circumstances, and comorbidities. Traditionally, the chronic kidney disease burden has been ascribed largely to hypertension and diabetes. Increasingly, evidence is accumulating that nontraditional risk factors may predominate in some regions and populations, contributing to epidemics of kidney disease. Such nontraditional risk factors include environmental exposures, traditional medicines, fetal and maternal factors, infections, kidney stones, and acute kidney injury. Genetic factors may predispose patients to chronic kidney disease in some populations. Chronic kidney disease of unknown origin has its epicenters in Central America and South Asia. Such clustering of CKD may represent either genetic or environmentally driven kidney disease, or combinations of both. Developmental conditions impacting kidney development often are related to poverty and structural factors that persist throughout life. In this article, we explore the possibilities that genetic and developmental factors may be important contributors to the epidemics in these regions and suggest that optimization of factors impacting kidney development hold promise to reduce the risk of kidney disease in future generations.
Collapse
Affiliation(s)
- David Friedman
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Valerie A Luyckx
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Institute for Biomedical Ethics and History of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
27
|
Shah SS, Lannon H, Dias L, Zhang JY, Alper SL, Pollak MR, Friedman DJ. APOL1 Kidney Risk Variants Induce Cell Death via Mitochondrial Translocation and Opening of the Mitochondrial Permeability Transition Pore. J Am Soc Nephrol 2019; 30:2355-2368. [PMID: 31558683 DOI: 10.1681/asn.2019020114] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 08/15/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Genetic Variants in Apolipoprotein L1 (APOL1) are associated with large increases in CKD rates among African Americans. Experiments in cell and mouse models suggest that these risk-related polymorphisms are toxic gain-of-function variants that cause kidney dysfunction, following a recessive mode of inheritance. Recent data in trypanosomes and in human cells indicate that such variants may cause toxicity through their effects on mitochondria. METHODS To examine the molecular mechanisms underlying APOL1 risk variant-induced mitochondrial dysfunction, we generated tetracycline-inducible HEK293 T-REx cells stably expressing the APOL1 nonrisk G0 variant or APOL1 risk variants. Using these cells, we mapped the molecular pathway from mitochondrial import of APOL1 protein to APOL1-induced cell death with small interfering RNA knockdowns, pharmacologic inhibitors, blue native PAGE, mass spectrometry, and assessment of mitochondrial permeability transition pore function. RESULTS We found that the APOL1 G0 and risk variant proteins shared the same import pathway into the mitochondrial matrix. Once inside, G0 remained monomeric, whereas risk variant proteins were prone to forming higher-order oligomers. Both nonrisk G0 and risk variant proteins bound components of the mitochondrial permeability transition pore, but only risk variant proteins activated pore opening. Blocking mitochondrial import of APOL1 risk variants largely eliminated oligomer formation and also rescued toxicity. CONCLUSIONS Our study illuminates important differences in the molecular behavior of APOL1 nonrisk and risk variants, and our observations suggest a mechanism that may explain the very different functional effects of these variants, despite the lack of consistently observed differences in trafficking patterns, intracellular localization, or binding partners. Variant-dependent differences in oligomerization pattern may underlie APOL1's recessive, gain-of-function biology.
Collapse
Affiliation(s)
- Shrijal S Shah
- Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Herbert Lannon
- Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Leny Dias
- Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jia-Yue Zhang
- Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Seth L Alper
- Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Martin R Pollak
- Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - David J Friedman
- Renal Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
28
|
|
29
|
Kamoto K, Noyes H, Nambala P, Senga E, Musaya J, Kumwenda B, Bucheton B, Macleod A, Cooper A, Clucas C, Herz-Fowler C, Matove E, Chiwaya AM, Chisi JE. Association of APOL1 renal disease risk alleles with Trypanosoma brucei rhodesiense infection outcomes in the northern part of Malawi. PLoS Negl Trop Dis 2019; 13:e0007603. [PMID: 31412021 PMCID: PMC6750591 DOI: 10.1371/journal.pntd.0007603] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/18/2019] [Accepted: 07/04/2019] [Indexed: 12/19/2022] Open
Abstract
Trypanosoma brucei (T.b.) rhodesiense is the cause of the acute form of human African trypanosomiasis (HAT) in eastern and southern African countries. There is some evidence that there is diversity in the disease progression of T.b. rhodesiense in different countries. HAT in Malawi is associated with a chronic haemo-lymphatic stage infection compared to other countries, such as Uganda, where the disease is acute with more marked neurological impairment. This has raised the question of the role of host genetic factors in infection outcomes. A candidate gene association study was conducted in the northern region of Malawi. This was a case-control study involving 202 subjects, 70 cases and 132 controls. All individuals were from one area; born in the area and had been exposed to the risk of infection since birth. Ninety-six markers were genotyped from 17 genes: IL10, IL8, IL4, HLA-G, TNFA, IL6, IFNG, MIF, APOL, HLA-A, IL1B, IL4R, IL12B, IL12R, HP, HPR, and CFH. There was a strong significant association with APOL1 G2 allele (p = 0.0000105, OR = 0.14, CI95 = [0.05-0.41], BONF = 0.00068) indicating that carriers of the G2 allele were protected against T.b. rhodesiense HAT. SNP rs2069845 in IL6 had raw p < 0.05, but did not remain significant after Bonferroni correction. There were no associations found with the other 15 candidate genes. Our finding confirms results from other studies that the G2 variant of APOL1 is associated with protection against T.b. rhodesiense HAT.
Collapse
Affiliation(s)
- Kelita Kamoto
- University of Malawi, College of Medicine, Department of Basic Medical Sciences, Blantyre, Malawi
| | - Harry Noyes
- Centre for Genomic Research, University of Liverpool, United Kingdom
| | - Peter Nambala
- University of Malawi, College of Medicine, Department of Basic Medical Sciences, Blantyre, Malawi
| | - Edward Senga
- University of Malawi, College of Medicine, Department of Basic Medical Sciences, Blantyre, Malawi
| | - Janelisa Musaya
- University of Malawi, College of Medicine, Department of Basic Medical Sciences, Blantyre, Malawi
| | - Benjamin Kumwenda
- University of Malawi, College of Medicine, Department of Basic Medical Sciences, Blantyre, Malawi
| | - Bruno Bucheton
- Institut de Recherche pour le Développement (IRD), IRD-CIRAD 177, Montpellier, France
- Programme National de Lutte contre la Trypanosomose Humaine Africaine, Conakry, Guinea
| | - Annette Macleod
- Wellcome Trust Centre for Molecular Parasitology, University Place, Glasgow, United Kingdom
| | - Anneli Cooper
- Wellcome Trust Centre for Molecular Parasitology, University Place, Glasgow, United Kingdom
| | - Caroline Clucas
- Wellcome Trust Centre for Molecular Parasitology, University Place, Glasgow, United Kingdom
| | | | | | | | - John E. Chisi
- University of Malawi, College of Medicine, Department of Basic Medical Sciences, Blantyre, Malawi
| | | |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) can cluster in geographic locations or in people of particular genetic ancestries. We explore APOL1 nephropathy and Balkan nephropathy as examples of CKD clustering that illustrate genetics and environment conspiring to cause high rates of kidney disease. Unexplained hotspots of kidney disease in Asia and Central America are then considered from the perspective of potential gene × environment interactions. RECENT FINDINGS We report on evidence supporting both genes and environment in these CKD hotspots. Differing genetic susceptibility between populations and within populations may explain why causal environmental risk factors have been so hard to identify conclusively. Similarly, one cannot explain why these epidemics of kidney disease are happening now without invoking environmental changes. SUMMARY Approaches to these CKD hotspots are of necessity becoming more holistic. Genetic studies may help us identify the environmental triggers by teaching us about disease biology and may empower environmental risk factor studies by allowing for stratification of study participants by genetic susceptibility.
Collapse
|
31
|
Musah S, Dimitrakakis N, Camacho DM, Church GM, Ingber DE. Directed differentiation of human induced pluripotent stem cells into mature kidney podocytes and establishment of a Glomerulus Chip. Nat Protoc 2019; 13:1662-1685. [PMID: 29995874 DOI: 10.1038/s41596-018-0007-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protocols have been established to direct the differentiation of human induced pluripotent stem (iPS) cells into nephron progenitor cells and organoids containing many types of kidney cells, but it has been difficult to direct the differentiation of iPS cells to form specific types of mature human kidney cells with high yield. Here, we describe a detailed protocol for the directed differentiation of human iPS cells into mature, post-mitotic kidney glomerular podocytes with high (>90%) efficiency within 26 d and under chemically defined conditions, without genetic manipulations or subpopulation selection. We also describe how these iPS cell-derived podocytes may be induced to form within a microfluidic organ-on-a-chip (Organ Chip) culture device to build a human kidney Glomerulus Chip that mimics the structure and function of the kidney glomerular capillary wall in vitro within 35 d (starting with undifferentiated iPS cells). The podocyte differentiation protocol requires skills for culturing iPS cells, and the development of a Glomerulus Chip requires some experience with building and operating microfluidic cell culture systems. This method could be useful for applications in nephrotoxicity screening, therapeutic development, and regenerative medicine, as well as mechanistic study of kidney development and disease.
Collapse
Affiliation(s)
- Samira Musah
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Diogo M Camacho
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA. .,Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. .,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
32
|
Gordon EJ, Wicklund C, Lee J, Sharp RR, Friedewald J. A National Survey of Transplant Surgeons and Nephrologists on Implementing Apolipoprotein L1 ( APOL1) Genetic Testing Into Clinical Practice. Prog Transplant 2019; 29:26-35. [PMID: 30541404 PMCID: PMC9527710 DOI: 10.1177/1526924818817048] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
INTRODUCTION There is debate over whether Apolipoprotein L1 (APOL1) gene risk variants contribute to African American (AA) live donors' (LD) increased risk of kidney failure. Little is known about factors influencing physicians' integration of APOL1 genetic testing of AA LDs into donor evaluation. DESIGN We conducted a cross-sectional survey, informed by Roger's Diffusion of Innovations theory, among nephrology and surgeon members of the American Society of Nephrology, American Society of Transplantation, and American Society of Transplant Surgeons about their practices of and attitudes about APOL1 genetic testing of AA potential LDs. Descriptive statistics and bivariate analyses were performed. RESULTS Of 383 completed surveys, most physicians believed that APOL1 testing can help AA LDs make more informed donation decisions (87%), and the addition of APOL1 testing offers better clinical information about AA LD's eligibility for donation than existing evaluation approaches (74%). Among respondents who evaluate LDs (n = 345), 63% would definitely or probably begin or continue using APOL1 testing in the next year, however, few use APOL1 testing routinely (4%) or on a case-by-case basis (14%). Most did not know the right clinical scenario to order APOL1 testing (59%), but would use educational materials to counsel AA LDs about APOL1 testing (97%). DISCUSSION Although physicians were highly supportive of APOL1 genetic testing for AA LDs, few physicians use APOL1 testing. As more physicians intend to use APOL1 testing, an ethical framework and clinical decision support are needed presently to assist clinicians in clarifying the proper indication of APOL1 genetic testing.
Collapse
Affiliation(s)
- Elisa J Gordon
- Division of Transplantation, Department of Surgery, Center for Healthcare Studies, Center for Bioethics and Medical Humanities, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Catherine Wicklund
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jungwha Lee
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Richard R Sharp
- Biomedical Ethics Research Program Mayo Clinic, Rochester, MN, USA
| | - John Friedewald
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
33
|
Lau RW, Wang B, Ricardo SD. Gene editing of stem cells for kidney disease modelling and therapeutic intervention. Nephrology (Carlton) 2019; 23:981-990. [PMID: 29851168 DOI: 10.1111/nep.13410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2018] [Indexed: 12/13/2022]
Abstract
Recent developments in targeted gene editing have paved the way for the wide adoption of clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein-9 nucleases (Cas9) as an RNA-guided molecular tool to modify the genome of eukaryotic cells of animals. Theoretically, the translation of CRISPR-Cas9 can be applied to the treatment of inherited or acquired kidney disease, kidney transplantation and genetic corrections of somatic cells from kidneys with inherited mutations, such as polycystic kidney disease. Human pluripotent stem cells have been used to generate an unlimited source of kidney progenitor cells or, when spontaneously differentiated into three-dimensional kidney organoids, to model kidney organogenesis or the pathogenesis of disease. Gene editing now allows for the tagging and selection of specific kidney cell types or disease-specific gene knock in/out, which enables more precise understanding of kidney organogenesis and genetic diseases. This review discusses the mechanisms of action, in addition to the advantages and disadvantages, of the three major gene editing technologies, namely, CRISPR-Cas9, zinc finger nucleases and transcription activator-like effector nucleases. The implications of using gene editing to better understand kidney disease is reviewed in detail. In addition, the ethical issues of gene editing, which could be easily neglected in the modern, fast-paced research environment, are highlighted.
Collapse
Affiliation(s)
- Ricky Wk Lau
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Bo Wang
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Suarez J, Cohen JB, Potluri V, Yang W, Kaplan DE, Serper M, Shah SP, Reese PP. Racial Disparities in Nephrology Consultation and Disease Progression among Veterans with CKD: An Observational Cohort Study. J Am Soc Nephrol 2018; 29:2563-2573. [PMID: 30120108 DOI: 10.1681/asn.2018040344] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/20/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Incident rates of ESRD are much higher among black and Hispanic patients than white patients. Access to nephrology care before progression to ESRD is associated with better clinical outcomes among patients with CKD. However, it is unknown whether black or Hispanic patients with CKD experience lower pre-ESRD nephrology consultation rates compared with their white counterparts, or whether such a disparity contributes to worse outcomes among minorities. METHODS We assembled a retrospective cohort of patients with CKD who received care through the Veterans Health Administration from 2003 to 2015, focusing on individuals with incident CKD stage 4 who had an initial eGFR≥60 ml/min per 1.73 m2 followed by two consecutive eGFRs<30 ml/min per 1.73 m2. We repeated analyses among individuals with incident CKD stage 3. Outcomes included nephrology provider referral, nephrology provider visit, progression to CKD stage 5, and mortality. RESULTS We identified 56,767 veterans with CKD stage 4 and 640,704 with CKD stage 3. In both cohorts, rates of nephrology referral and visits were significantly higher among black and Hispanic veterans than among non-Hispanic white veterans. Despite this, both black and Hispanic patients experienced faster progression to CKD stage 5 compared with white patients. Black patients with CKD stage 4 experienced slightly lower mortality than white patients, whereas black patients with CKD stage 3 had a small increased risk of death. CONCLUSIONS Black or Hispanic veterans with CKD are more likely than white patients to see a nephrologist, yet are also more likely to suffer disease progression. Biologic and environmental factors may play a bigger role than nephrology consultation in driving racial disparities in CKD progression.
Collapse
Affiliation(s)
- Jonathan Suarez
- Renal-Electrolyte and Hypertension Division, Department of Medicine, and
| | - Jordana B Cohen
- Renal-Electrolyte and Hypertension Division, Department of Medicine, and.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vishnu Potluri
- Renal-Electrolyte and Hypertension Division, Department of Medicine, and
| | - Wei Yang
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - David E Kaplan
- Gastroenterology Section, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania; and.,Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marina Serper
- Gastroenterology Section, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania; and.,Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Siddharth P Shah
- Renal-Electrolyte and Hypertension Division, Department of Medicine, and
| | - Peter Philip Reese
- Renal-Electrolyte and Hypertension Division, Department of Medicine, and .,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Gastroenterology Section, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania; and
| |
Collapse
|
35
|
Siemens TA, Riella MC, Moraes TPD, Riella CV. APOL1 risk variants and kidney disease: what we know so far. ACTA ACUST UNITED AC 2018; 40:388-402. [PMID: 30052698 PMCID: PMC6533999 DOI: 10.1590/2175-8239-jbn-2017-0033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/19/2018] [Indexed: 01/08/2023]
Abstract
There are striking differences in chronic kidney disease between Caucasians and African descendants. It was widely accepted that this occurred due to socioeconomic factors, but recent studies show that apolipoprotein L-1 (APOL1) gene variants are strongly associated with focal segmental glomerulosclerosis, HIV-associated nephropathy, hypertensive nephrosclerosis, and lupus nephritis in the African American population. These variants made their way to South America trough intercontinental slave traffic and conferred an evolutionary advantage to the carries by protecting against forms of trypanosomiasis, but at the expense of an increased risk of kidney disease. The effect of the variants does not seem to be related to their serum concentration, but rather to local action on the podocytes. Risk variants are also important in renal transplantation, since grafts from donors with risk variants present worse survival.
Collapse
|
36
|
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
|
37
|
Gutiérrez OM, Irvin MR, Chaudhary NS, Cushman M, Zakai NA, David VA, Limou S, Pamir N, Reiner AP, Naik RP, Sale MM, Safford MM, Hyacinth HI, Judd SE, Kopp JB, Winkler CA. APOL1 Nephropathy Risk Variants and Incident Cardiovascular Disease Events in Community-Dwelling Black Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e002098. [PMID: 29899045 PMCID: PMC6339526 DOI: 10.1161/circgen.117.002098] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/04/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND APOL1 renal risk variants are strongly associated with chronic kidney disease in Black adults, but reported associations with cardiovascular disease (CVD) have been conflicting. METHODS We examined associations of APOL1 with incident coronary heart disease (n=323), ischemic stroke (n=331), and the composite CVD outcome (n=500) in 10 605 Black participants of the REGARDS study (Reasons for Geographic and Racial Differences in Stroke). Primary analyses compared individuals with APOL1 high-risk genotypes to APOL1 low-risk genotypes in Cox proportional hazards models adjusted for CVD risk factors and African ancestry. RESULTS APOL1 high-risk participants were younger and more likely to have albuminuria at baseline than APOL1 low-risk participants. The risk of incident stroke, coronary heart disease, or composite CVD end point did not significantly differ by APOL1 genotype status in multivariable models. The association of APOL1 genotype with incident composite CVD differed by diabetes mellitus status (Pinteraction=0.004). In those without diabetes mellitus, APOL1 high-risk genotypes associated with greater risk of incident composite CVD (hazard ratio, 1.67; 95% confidence interval, 1.12-2.47) compared with those with APOL1 low-risk genotypes in multivariable adjusted models. This latter association was driven by ischemic strokes (hazard ratio, 2.32; 95% confidence interval, 1.33-4.07), in particular, those related to small vessel disease (hazard ratio, 5.10; 95% confidence interval, 1.55-16.56). There was no statistically significant association of APOL1 genotypes with incident CVD in subjects with diabetes mellitus. The APOL1 high-risk genotype was associated with higher stroke risk in individuals without but not those with chronic kidney disease in fully adjusted models. CONCLUSIONS APOL1 high-risk status is associated with CVD events in community-dwelling Black adults without diabetes mellitus.
Collapse
Affiliation(s)
- Orlando M Gutiérrez
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL (O.M.G.)
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL (M.R.I., N.S.C.)
| | - Ninad S Chaudhary
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL (M.R.I., N.S.C.)
| | - Mary Cushman
- Division of Hematology and Oncology, University of Vermont, Burlington, VT (M.C., N.A.Z.)
| | - Neil A Zakai
- Division of Hematology and Oncology, University of Vermont, Burlington, VT (M.C., N.A.Z.)
| | - Victor A David
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD (V.A.D.)
| | - Sophie Limou
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD (S.L., C.A.W.)
- Center for Research in Immunology and Transplantation, University of Nantes, Nantes, France (S.L.)
| | - Nathalie Pamir
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR (N.P.)
| | - Alex P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA (A.P.R.)
| | - Rakhi P Naik
- Department of Medicine, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD (R.P.N.)
| | - Michele M Sale
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA (M.M. Sale)
| | - Monika M Safford
- Division of General Internal Medicine, Weill Cornell Medicine, New York, NY (M.M. Safford)
| | - Hyacinth I Hyacinth
- Department of Pediatrics, Aflac Cancer and Blood Disorder Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA (H.I.H.)
| | - Suzanne E Judd
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL (S.E.J.)
| | - Jeffrey B Kopp
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.B.K.).
| | - Cheryl A Winkler
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD (S.L., C.A.W.)
| |
Collapse
|
38
|
Locke JE, Sawinski D, Reed RD, Shelton B, MacLennan PA, Kumar V, Mehta S, Mannon RB, Gaston R, Julian BA, Carr JJ, Terry JG, Kilgore M, Massie AB, Segev DL, Lewis CE. Apolipoprotein L1 and Chronic Kidney Disease Risk in Young Potential Living Kidney Donors. Ann Surg 2018; 267:1161-1168. [PMID: 28187045 PMCID: PMC5805656 DOI: 10.1097/sla.0000000000002174] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The aim of this study was to develop a novel chronic kidney disease (CKD) risk prediction tool for young potential living kidney donors. SUMMARY OF BACKGROUND DATA Living kidney donor selection practices have evolved from examining individual risk factors to a risk calculator incorporating multiple characteristics. Owing to limited long-term data and lack of genetic information, current risk tools lack precision among young potential living kidney donors, particularly African Americans (AAs). METHODS We identified a cohort of young adults (18-30 years) with no absolute contraindication to kidney donation from the longitudinal cohort study Coronary Artery Risk Development in Young Adults. Risk associations for CKD (estimated glomerular filtration rate <60 mL/min/1.73 m) were identified and assigned weighted points to calculate risk scores. RESULTS A total of 3438 healthy adults were identified [mean age 24.8 years; 48.3% AA; median follow-up 24.9 years (interquartile range: 24.5-25.2)]. For 18-year olds, 25-year projected CKD risk varied by ethnicity and sex even without baseline clinical and genetic abnormalities; risk was 0.30% for European American (EA) women, 0.52% for EA men, 0.52% for AA women, 0.90% for AA men. Among 18-year-old AAs with apolipoprotein L1 gene (APOL1) renal-risk variants without baseline abnormalities, 25-year risk significantly increased: 1.46% for women and 2.53% for men; among those with 2 APOL1 renal-risk variants and baseline abnormalities, 25-year risk was higher: 2.53% to 6.23% for women and 4.35% to 10.58% for men. CONCLUSIONS Young AAs were at highest risk for CKD, and APOL1 renal-risk variants drove some of this risk. Understanding the genetic profile of young AA potential living kidney donors in the context of baseline health characteristics may help to inform candidate selection and counseling.
Collapse
Affiliation(s)
- Jayme E. Locke
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Deirdre Sawinski
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Rhiannon D. Reed
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Brittany Shelton
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Paul A. MacLennan
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Vineeta Kumar
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shikha Mehta
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Roslyn B. Mannon
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert Gaston
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bruce A. Julian
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - J. Jeffrey Carr
- Department of Radiology and Vanderbilt Translational and Clinical Cardiovascular Research Center (VTRACC), Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James G. Terry
- Department of Radiology and Vanderbilt Translational and Clinical Cardiovascular Research Center (VTRACC), Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Meredith Kilgore
- University of Alabama at Birmingham School of Public Health, Birmingham, Alabama
| | | | - Dorry L. Segev
- Johns Hopkins University School of Medicine, Baltimore, MD
| | - Cora E. Lewis
- University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| |
Collapse
|
39
|
Ethnicity matching and outcomes after kidney transplantation in the United Kingdom. PLoS One 2018; 13:e0195038. [PMID: 29652887 PMCID: PMC5898720 DOI: 10.1371/journal.pone.0195038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/15/2018] [Indexed: 01/13/2023] Open
Abstract
Background Kidneys from non-white donors have inferior outcomes, but it is unclear if ethnicity matching between donors and recipients achieves better post kidney transplant outcomes. Methods We undertook a retrospective, population cohort study utilising UK Transplant Registry data. The cohort comprised adult, kidney-alone, transplant recipients receiving their first kidney transplant between 2003–2015, with data censored at 1st October 2016. We included 27,970 recipients stratified into white (n = 23,215), black (n = 1,679) and south Asian (n = 3,076) ethnicity, with median post-transplant follow-up of 1,676 days (IQR 716–2,869 days). Unadjusted and adjusted Cox regression survival analyses were performed to investigate ethnicity effect on risk for graft loss and mortality. Results In unadjusted analyses, matched ethnicity between donors-recipients resulted in better outcomes for delayed graft function, one-year creatinine, graft and patient survival but these differed by ethnicity matches. Compared to white-to-white transplants, risk for death-censored graft loss was higher in black-to-black and similar among Asian-to-Asian transplants, but mortality risk was lower for both black-to-black and Asian-to-Asian transplants. In Cox regression models, compared to white donors, we observed higher risk for graft loss with both south Asian (HR 1.38, 95%CI 1.12–1.70, p = 0.003) and black (HR 1.66, 95%CI 1.30–2.11, p<0.001) donated kidneys independent of recipient ethnicity. We observed no mortality difference with south Asian donated kidneys but increased mortality with black donated kidneys (HR 1.68, 95%CI 1.21–2.35, p = 0.002). Matching ethnicities made no significant difference in any Cox regression model. Similar results were observed after stratifying our analysis by living and deceased-donor kidney transplantation. Conclusions Our data confirm inferior outcomes associated with non-white kidney donors for kidney transplant recipients of any ethnicity in a risk-adjusted model for the United Kingdom population. However, contrary to non-renal transplant literature, we did not identify any survival benefits associated with donor-recipient ethnicity matching.
Collapse
|
40
|
Guan M, Keaton JM, Dimitrov L, Hicks PJ, Xu J, Palmer ND, Wilson JG, Freedman BI, Bowden DW, Ng MC. An Exome-wide Association Study for Type 2 Diabetes-Attributed End-Stage Kidney Disease in African Americans. Kidney Int Rep 2018; 3:867-878. [PMID: 29989002 PMCID: PMC6035163 DOI: 10.1016/j.ekir.2018.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/20/2018] [Accepted: 03/05/2018] [Indexed: 12/12/2022] Open
Abstract
Introduction Compared with European Americans, African Americans (AAs) are at higher risk for developing end-stage kidney disease (ESKD). Genome-wide association studies (GWAS) have identified >70 genetic variants associated with kidney function and chronic kidney disease (CKD) in patients with and without diabetes. However, these variants explain a small proportion of disease liability. This study examined the contribution of coding genetic variants for risk of type 2 diabetes (T2D)-attributed ESKD and advanced CKD in AAs. Methods Exome sequencing was performed in 456 AA T2D-ESKD cases, and 936 AA nondiabetic, non-nephropathy control individuals at the discovery stage. A mixed logistic regression model was used for association analysis. Nominal associations (P < 0.05) were replicated in an additional 2020 T2D-ESKD cases and 1121 nondiabetic, non-nephropathy control individuals. A meta-analysis combining 4533 discovery and replication samples was performed. Putative T2D-ESKD associations were tested in additional 1910 nondiabetic ESKD and 219 T2D-ESKD cases, as well as 912 AA nondiabetic non-nephropathy control individuals. Results A total of 11 suggestive T2D-ESKD associations (P < 1 x 10−4) from 8 loci (PLEKHN1, NADK, RAD51AP2, RREB1, PEX6, GRM8, PRX, APOL1) were apparent in the meta-analysis. Exclusion of APOL1 renal-risk genotype carriers identified 3 additional suggestive loci (OTUD7B, IFITM3, DLGAP5). Rs41302867 in RREB1 displayed consistent association with T2D-ESKD and nondiabetic ESKD (odds ratio: 0.47; P = 1.2 x 10−6 in 4605 all-cause ESKD and 2969 nondiabetic non-nephropathy control individuals). Conclusion Our findings suggest that coding genetic variants are implicated in predisposition to T2D-ESKD in AAs.
Collapse
Affiliation(s)
- Meijian Guan
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jacob M. Keaton
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Latchezar Dimitrov
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Pamela J. Hicks
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jianzhao Xu
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Nicholette D. Palmer
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Barry I. Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Donald W. Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Maggie C.Y. Ng
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Correspondence: Maggie C. Y. Ng, Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| |
Collapse
|
41
|
Affiliation(s)
- Martin Pollak
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA.
| |
Collapse
|
42
|
Horowitz CR, Ferryman K, Negron R, Sabin T, Rodriguez M, Zinberg RF, Böttinger E, Robinson M. Race, Genomics and Chronic Disease: What Patients with African Ancestry Have to Say. J Health Care Poor Underserved 2018; 28:248-260. [PMID: 28238999 DOI: 10.1353/hpu.2017.0020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Variants of the APOL1 gene increase risk for kidney failure 10-fold, and are nearly exclusively found in people with African ancestry. To translate genomic discoveries into practice, we gathered information about effects and challenges incorporating genetic risk in clinical care. METHODS An academic-community-clinical team tested 26 adults with self-reported African ancestry for APOL1 variants, conducting in-depth interviews about patients' beliefs and attitudes toward genetic testing- before, immediately, and 30 days after receiving test results. We used constant comparative analysis of interview transcripts to identify themes. RESULTS Themes included: Knowledge of genetic risk for kidney failure may motivate providers and patients to take hypertension more seriously, rather than inspiring fatalism or anxiety. Having genetic risk for a disease may counter stereotypes of Blacks as non-adherent or low-literate, rather than exacerbate stereotypes. CONCLUSION Populations most likely to benefit from genomic research can inform strategies for genetic testing and future research.
Collapse
|
43
|
Rhee EP. How Omics Data Can Be Used in Nephrology. Am J Kidney Dis 2018; 72:129-135. [PMID: 29478865 DOI: 10.1053/j.ajkd.2017.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/10/2017] [Indexed: 02/07/2023]
Abstract
Advances in technology and computing now permit the high-throughput analysis of multiple domains of biological information, including the genome, transcriptome, proteome, and metabolome. These omics approaches, particularly comprehensive analysis of the genome, have catalyzed major discoveries in science and medicine, including in nephrology. However, they also generate large complex data sets that can be difficult to synthesize from a clinical perspective. This article seeks to provide an overview that makes omics technologies relevant to the practicing nephrologist, framing these tools as an extension of how we approach patient care in the clinic. More specifically, omics technologies reinforce the impact that genetic mutations can have on a range of kidney disorders, expand the catalogue of uremic molecules that accumulate in blood with kidney failure, enhance our ability to scrutinize urine beyond urinalysis for insight on renal pathology, and enable more extensive characterization of kidney tissue when a biopsy is performed. Although assay methodologies vary widely, all omics technologies share a common conceptual framework that embraces unbiased discovery at the molecular level. Ultimately, the application of these technologies seeks to elucidate a more mechanistic and individualized approach to the diagnosis and treatment of human disease.
Collapse
Affiliation(s)
- Eugene P Rhee
- Nephrology and Endocrinology Divisions, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
44
|
Liu YC, Chun J. Prospects for Precision Medicine in Glomerulonephritis Treatment. Can J Kidney Health Dis 2018; 5:2054358117753617. [PMID: 29449955 PMCID: PMC5808958 DOI: 10.1177/2054358117753617] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/12/2017] [Indexed: 02/06/2023] Open
Abstract
Background: Glomerulonephritis (GN) consists of a group of kidney diseases that are categorized based on shared histopathological features. The current classifications for GN make it difficult to distinguish the individual variability in presentation, disease progression, and response to treatment. GN is a significant cause of end-stage renal disease (ESRD), and improved therapies are desperately needed because current immunosuppressive therapies sometimes lack efficacy and can lead to significant toxicities. In recent years, the combination of high-throughput genetic approaches and technological advances has identified important regulators contributing to GN. Objectives: In this review, we summarize recent findings in podocyte biology and advances in experimental approaches that have opened the possibility of precision medicine in GN treatment. We provide an integrative basic science and clinical overview of new developments in GN research and the discovery of potential candidates for targeted therapies in GN. Findings: Advances in podocyte biology have identified many candidates for therapeutic targets and potential biomarkers of glomerular disease. The goal of precision medicine in GN is now being pursued with recent technological improvements in genetics, accessibility of biologic and clinical information with tissue biobanks, high-throughput analysis of large-scale data sets, and new human model systems such as kidney organoids. Conclusion: With advances in data collection, technologies, and experimental model systems, we now have vast tools available to pursue precision medicine in GN. We anticipate a growing number of studies integrating data from high-throughput analysis with the development of diagnostic tools and targeted therapies for GN in the near future.
Collapse
Affiliation(s)
- Yulu Cherry Liu
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Justin Chun
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Division of Nephrology, Department of Medicine, University of Calgary, Alberta, Canada
| |
Collapse
|
45
|
Kwan JM, Hajjiri Z, Chen YF, Metwally A, Perkins DL, Finn PW. Donor and Recipient Ethnicity Impacts Renal Graft Adverse Outcomes. J Racial Ethn Health Disparities 2017; 5:1003-1013. [DOI: 10.1007/s40615-017-0447-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/20/2017] [Accepted: 10/30/2017] [Indexed: 11/30/2022]
|
46
|
Nel M, Buys JM, Botha FCJ, Wearne N, Prince S, Heckmann JM. The functionality of African-specific variants in the TGFB1 regulatory region and their potential role in HIVAN. Clin Exp Nephrol 2017; 22:764-772. [PMID: 29204904 DOI: 10.1007/s10157-017-1516-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 11/23/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND Transcription of transforming growth factor beta-1 (TGF-β1) is regulated by a polymorphic promoter region containing African-specific single nucleotide polymorphisms (SNPs). Some of these SNPs have higher frequencies among Southern Africans compared to other African populations and their functionality has only been partially studied. Due to the high prevalence of HIV-associated nephropathy (HIVAN) in Africans we hypothesized that functional African TGFB1-promoter SNPs may contribute to HIVAN pathogenesis. METHODS The functionality of the TGFB1 -1347 C>T variant and African-specific variants (-1287 G>A, -1154 C>T, -387 C>T and -14 G>A) were examined by measuring reporter gene expression in kidney and fibroblast cell lines co-transfected with TGFB1-promoter constructs and an HIV-Tat expression vector. TGF-β1 immunohistochemical staining was performed on kidney biopsies with HIVAN (n = 18) and compared to control biopsies without HIVAN or tubulointerstitial disease (n = 12) using semi-quantitative and digital image analysis. HIVAN cases were genotyped for TGFB1 -1347 and -387 SNP variants. RESULTS TGFB1-promoter haplotypes containing the African -387 T-allele resulted in ~ five-fold repression of TGFB1-promoter activity compared to -387 C haplotypes (p ≤ 0.024). HIV-Tat upregulated TGFB1-promoter activity for haplotypes containing -1347 T and -387 T in transfected renal cells (≈ 1.6-fold; p ≤ 0.030) and fibroblasts (≈ 1.3-fold; p ≤ 0.016). The renal interstitium from HIVAN biopsies, compared to HIV-positive and -negative controls, differed in the semi-quantitative TGF-β1 staining and digital optical density analyses. The TGFB1 -1347 and -387 genotypes in HIVAN cases were similar to population controls. CONCLUSION African-specific haplotypes lower TGFB1-promoter activity and expression levels and HIV-Tat upregulates TGFB1 promoter activity irrespective of the haplotype.
Collapse
Affiliation(s)
- M Nel
- Neurology Research Group, Division of Neurology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - J-M Buys
- Neurology Research Group, Division of Neurology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - F C J Botha
- Division of Anatomical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - N Wearne
- Division of Nephrology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - S Prince
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - J M Heckmann
- Neurology Research Group, Division of Neurology, Department of Medicine, University of Cape Town, Cape Town, South Africa.
- Division of Neurology, Department of Medicine, Groote Schuur Hospital, Observatory, Cape Town, 7925, South Africa.
| |
Collapse
|
47
|
|
48
|
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
|
49
|
Pattaro C. Genome-wide association studies of albuminuria: towards genetic stratification in diabetes? J Nephrol 2017; 31:475-487. [PMID: 28918587 DOI: 10.1007/s40620-017-0437-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 09/02/2017] [Indexed: 12/16/2022]
Abstract
Genome-wide association studies (GWAS) have been very successful in unraveling the polygenic structure of several complex diseases and traits. In the case of albuminuria, despite the large sample size achieved by some studies, results look sparse with a limited number of loci reported so far. This review searched for GWAS studies of albumin excretion, albuminuria, and proteinuria. The resulting picture sets elements of uniqueness for albuminuria GWAS with respect to other complex traits. So far, very few loci associated with albuminuria have been validated by means of genome-wide significant evidence or formal replication. With rare exceptions, the validated loci are ethnicity specific. Within a given ethnicity, variants are common and have relatively large effects, especially in the presence of diabetes. In most cases, the identified variants were functional and a biological involvement of the target genes in renal damage was established. Recently reported variants associated with albuminuria in diabetes may be potentially combined into a genetic risk score, making it possible to rank diabetic patients by increasing risk of albuminuria. Validation of this model is required. To expand the understanding of the biological basis of albumin excretion regulation, future initiatives should achieve larger sample sizes and favor a transethnic study design.
Collapse
Affiliation(s)
- Cristian Pattaro
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100, Bolzano, Italy.
| |
Collapse
|
50
|
Harding K, Mersha TB, Webb FA, Vassalotti JA, Nicholas SB. Current State and Future Trends to Optimize the Care of African Americans with End-Stage Renal Disease. Am J Nephrol 2017; 46:156-164. [PMID: 28787724 DOI: 10.1159/000479479] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Chronic kidney disease is a progressive disease, which terminates in end-stage renal diseases (ESRD) that requires either dialysis or kidney transplantation for the patient to survive. There is an alarming trend in the disparities of ESRD in African Americans (AAs). Currently, AAs represent more than 30% of incident ESRD cases, yet they constitute 15% of the overall US population. Despite the reductions in mortality, increases in access to patient-centered home dialysis and preemptive kidney transplantation for the overall US ESRD population over the last decade, disparities in the care of AAs with ESRD remain largely unaffected. SUMMARY This review discusses patient-, community-, and practitioner-related factors that contribute to disparities in ESRD care for AAs. In particular, the review addresses issues related to end-of-life support, the importance of Apolipoprotein-1 gene variants, and the advent of pharmacogenomics toward achieving precision care. The need for accessible clinical intelligence for the ESRD population is discussed. Several interventions and a call to action to address the disparities are presented. Key Messages: Significant disparities in ESRD care exist for AAs. Strategies to enhance patient engagement, education, accountable partnerships, and clinical intelligence may reduce these disparities.
Collapse
|