1
|
Simeone CA, McNulty MT, Gupta Y, Genovese G, Sampson MG, Sanna-Cherchi S, Friedman DJ, Pollak MR. The APOL1 p.N264K variant is co-inherited with the G2 kidney disease risk variant through a proximity recombination event. G3 (BETHESDA, MD.) 2025; 15:jkae290. [PMID: 39658338 PMCID: PMC11797048 DOI: 10.1093/g3journal/jkae290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024]
Abstract
Black Americans are 3-4 times more likely to develop nondiabetic kidney disease than other populations. Exclusively found in people of recent African (AFR) ancestry, risk variants in Apolipoprotein L1 (APOL1) termed G1 and G2 contribute significantly to this increased susceptibility. Our group and others showed that a missense variant in APOL1, rs73885316 (p.N264K, "M1"), is remarkably protective against APOL1 kidney disease when co-inherited with the G2 risk allele. Since the distance between the M1 and G2 variants is only 367 base pairs, we initially suspected that 2 independent mutation events occurred to create non-risk M1-G0 and M1-G2 haplotypes. Here, we examined APOL1 haplotypes in individuals of AFR ancestry from the 1000 Genomes Project, the Nephrotic Syndrome Study Network (NEPTUNE), and an ancient individual from the Allen Ancient Genome Diversity Project to determine how the M1-G2 haplotype arose. We demonstrate that M1 most likely first appeared on a non-risk G0 haplotype, and that a subsequent recombination event bypassed strong recombination hotspots flanking APOL1 and occurred between p.N388Y389del on a G2 haplotype and M1 on a G0 haplotype to create the M1-G2 haplotype. Observing a recombination event within a small region between clinically relevant loci emphasizes the importance of studying the entire haplotype repertoire of a disease gene and the impact of haplotype backgrounds in disease susceptibility.
Collapse
Affiliation(s)
- Christopher A Simeone
- Harvard Medical School, Boston, MA 02215, USA
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Michelle T McNulty
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA 02215, USA
- Kidney Disease Initiative and Medical and Population Genetics Program, Broad Institute, Cambridge, MA 02142, USA
| | - Yask Gupta
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, Columbia University, New York City, NY 10032, USA
| | - Giulio Genovese
- Harvard Medical School, Boston, MA 02215, USA
- Stanley Center, Broad Institute of MIT and Harvard, Boston, MA 02215, USA
| | - Matthew G Sampson
- Harvard Medical School, Boston, MA 02215, USA
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA 02215, USA
- Kidney Disease Initiative and Medical and Population Genetics Program, Broad Institute, Cambridge, MA 02142, USA
- Division of Nephrology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, Columbia University, New York City, NY 10032, USA
| | - David J Friedman
- Harvard Medical School, Boston, MA 02215, USA
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Martin R Pollak
- Harvard Medical School, Boston, MA 02215, USA
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| |
Collapse
|
2
|
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
|
3
|
Ames EG, Anand PM, Bekheirnia MR, Doshi MD, El Ters M, Freese ME, Gbadegesin RA, Guay-Woodford LM, Java A, Ranch D, Rodig NM, Wang X, Thomas CP. Evaluation for genetic disease in kidney transplant candidates: A practice resource. Am J Transplant 2025; 25:237-249. [PMID: 39488252 DOI: 10.1016/j.ajt.2024.10.019] [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: 08/17/2024] [Revised: 10/08/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
The increasing availability of clinically approved genetic tests for kidney disease has spurred the growth in the use of these tests in kidney transplant practice. Neither the testing options nor the patient population where this should be deployed has been defined, and its value in kidney transplant evaluation has not been demonstrated. Transplant providers may not always be aware of the limitations of genetic testing and may need guidance on comprehending test results and providing counsel, as many centers do not have easy access to a renal genetic counselor or a clinical geneticist. In this practice resource, a working group of nephrologists, geneticists, and a genetic counselor provide a pragmatic, tailored approach to genetic testing, advocating for its use only where the genetic diagnosis or its exclusion can impact the choices available for transplantation or posttransplant management or the workup of living donor candidates at increased risk for heritable disease.
Collapse
Affiliation(s)
- Elizabeth G Ames
- Division of Pediatric Genetics, Metabolism, and Genomic Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Prince M Anand
- Department of Internal Medicine, Medical University of South Carolina, Lancaster, South Carolina, USA
| | - Mir Reza Bekheirnia
- Departments of Molecular and Human Genetics and Pediatrics, Baylor College of Medicine, Houston, Texas, USA; Michael E. Debakey VA Medical Center, Houston, Texas, USA
| | - Mona D Doshi
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mireille El Ters
- Division of Nephrology, Department of Medicine, William von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA
| | - Margaret E Freese
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Rasheed A Gbadegesin
- Division of Nephrology, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lisa M Guay-Woodford
- Divisions of Nephrology and Genetics, Research Institute and Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Anuja Java
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel Ranch
- Department of Pediatrics, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Nancy M Rodig
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Xiangling Wang
- Center for Personalized Genetic Healthcare, Department of Kidney Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Christie P Thomas
- Division of Nephrology, Department of Medicine, William von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA; Department of Internal Medicine, VA Medical Center, Iowa City, Iowa, USA.
| |
Collapse
|
4
|
Ben-Ruby D, Atias-Varon D, Kagan M, Chowers G, Shlomovitz O, Slabodnik-Kaner K, Mano N, Avayou S, Atsmony Y, Levin D, Dotan E, Calderon-Margalit R, Shnaider A, Haviv YS, Birk OS, Hadar N, Anikster Y, Berar Yanay N, Chernin G, Kruzel-Davila E, Beckerman P, Rozen-Zvi B, Doctor GT, Stanescu HC, Shemer R, Pras E, Reznik-Wolf H, Nahum AH, Dominissini D, Skorecki K, Vivante A. Multiethnic prevalence of the APOL1 G1 and G2 variants among the Israeli dialysis population. Clin Kidney J 2025; 18:sfae397. [PMID: 39927257 PMCID: PMC11803305 DOI: 10.1093/ckj/sfae397] [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: 10/08/2024] [Indexed: 02/11/2025] Open
Abstract
Background and hypothesis The two apolipoprotein L1 (APOL1) variants, G1 and G2, are common in populations of sub-Saharan African ancestry. Individuals with two of these alleles (G1 or G2) have an increased risk for a spectrum of non-diabetic chronic kidney diseases. However, these variants are typically not observed outside of populations that self-identify as current continental Africans or having clear recent African ancestry such as, most notably, African Americans, and other large population groups in the Americas and several European countries. We hypothesized that the diverse ethnic groups within the Israeli population may exhibit varying levels of recent African ancestry. Therefore, it is plausible that APOL1 risk alleles might be present even in individuals who do not self-identify as being of sub-Saharan African descent. Methods We non-selectively screened people with kidney failure across Israel for APOL1 risk variants using restriction fragment length polymorphism. Results We recruited 1744 individuals from 38 dialysis units in Israel. We identified eight patients of Moroccan Jewish, Bedouin, or Muslim Arab ancestry, who carry at least one G1 or G2 allele. None of the eight patients carried the protective APOL1 p.N264K variant. Furthermore, despite all Bedouin individuals being G2 heterozygous, the G2 minor allele frequency was significantly enriched in kidney failure cases compared to ethnically matched controls (P = .006). Conclusions These findings show that APOL1 G1 and G2 allelic variants are present in populations previously not appreciated to possess recent sub-Saharan ancestry and suggest that a single G2 risk variant may confer increased risk for chronic kidney disease in certain population contexts.
Collapse
Affiliation(s)
- Dror Ben-Ruby
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Danit Atias-Varon
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Maayan Kagan
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Guy Chowers
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Omer Shlomovitz
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Keren Slabodnik-Kaner
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Neta Mano
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Arrow Project, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Shany Avayou
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Yariv Atsmony
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Dana Levin
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Edo Dotan
- The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
- The Taub Faculty of Computer Science, Technion Israel Institute of Technology, Haifa, Israel
| | - Ronit Calderon-Margalit
- Braun School of Public Health, Hadassah Medical Center, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Alla Shnaider
- Department of Nephrology, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
| | - Yosef S Haviv
- Department of Nephrology, Soroka University Medical Center, Beer-Sheva, Israel
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
| | - Ohad S Birk
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
- Genetics Institute at Soroka Medical Center, Beer-Sheva, Israel
| | - Noam Hadar
- Faculty of Health Sciences, Ben Gurion University, Beer-Sheva, Israel
| | - Yair Anikster
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Metabolic Diseases Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Noa Berar Yanay
- Nephrology Department, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Gil Chernin
- Department of Nephrology and Hypertension, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Etty Kruzel-Davila
- Nephrology Department, Galilee Medical Center, Nahariya, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Pazit Beckerman
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Institute of Nephrology and Hypertension, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Benaya Rozen-Zvi
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Nephrology and Hypertension, Rabin Medical Center, Petah Tikva, Israel
| | - Gabriel T Doctor
- Centre for Genetics and Genomics, Department of Renal Medicine, UCL Division of Medicine, University College London, London, UK
| | - Horia C Stanescu
- Centre for Genetics and Genomics, Department of Renal Medicine, UCL Division of Medicine, University College London, London, UK
| | - Revital Shemer
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Elon Pras
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Haike Reznik-Wolf
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Ayelet Hashahar Nahum
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Dan Dominissini
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Institute of Hematology, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Karl Skorecki
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Rambam Health Care Campus, Haifa, Israel
| | - Asaf Vivante
- Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
- Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| |
Collapse
|
5
|
Gbadegesin RA, Ulasi I, Ajayi S, Raji Y, Olanrewaju T, Osafo C, Ademola AD, Asinobi A, Winkler CA, Burke D, Arogundade F, Ekem I, Plange-Rhule J, Mamven M, Matekole M, Amodu O, Cooper R, Antwi S, Adeyemo AA, Ilori TO, Adabayeri V, Nyarko A, Ghansah A, Amira T, Solarin A, Awobusuyi O, Kimmel PL, Brosius FC, Makusidi M, Odenigbo U, Kretzler M, Hodgin JB, Pollak MR, Boima V, Freedman BI, Palmer ND, Collins B, Phadnis M, Smith J, Agwai CI, Okoye O, Abdu A, Wilson J, Williams W, Salako BL, Parekh RS, Tayo B, Adu D, Ojo A. APOL1 Bi- and Monoallelic Variants and Chronic Kidney Disease in West Africans. N Engl J Med 2025; 392:228-238. [PMID: 39465900 PMCID: PMC11735277 DOI: 10.1056/nejmoa2404211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
BACKGROUND Apolipoprotein L1 gene (APOL1) variants are risk factors for chronic kidney disease (CKD) among Black Americans. Data are sparse on the genetic epidemiology of CKD and the clinical association of APOL1 variants with CKD in West Africans, a major group in the Black population. METHODS We conducted a case-control study involving participants from Ghana and Nigeria who had CKD stages 2 through 5, biopsy-proven glomerular disease, or no kidney disease. We analyzed the association of CKD with APOL1 variants among participants with high-risk genotypes (two APOL1 risk alleles) and those with low-risk genotypes (fewer than two APOL1 risk alleles) by fitting logistic-regression models that controlled for covariates, including clinical site, age, and sex. RESULTS Among 8355 participants (4712 with CKD stages 2 through 5, 866 with glomerular diseases, and 2777 with no kidney disease), the prevalence of monoallelic APOL1 variants was 43.0% and that of biallelic APOL1 variants was 29.7%. Participants with two APOL1 risk alleles had higher odds of having CKD than those with one risk allele or no risk alleles (adjusted odds ratio, 1.25; 95% confidence interval [CI], 1.11 to 1.40), as well as higher odds of focal segmental glomerulosclerosis (adjusted odds ratio, 1.84; 95% CI, 1.30 to 2.61). Participants with one APOL1 risk allele had higher odds of having CKD than those with no risk alleles (adjusted odds ratio, 1.18; 95% CI, 1.04 to 1.33), as well as higher odds of focal segmental glomerulosclerosis (adjusted odds ratio, 1.61; 95% CI, 1.04 to 2.48). The inclusion of covariates did not modify the association of monoallelic and biallelic APOL1 variants with CKD or focal segmental glomerulosclerosis. CONCLUSIONS In this study, monoallelic APOL1 variants were associated with 18% higher odds of CKD and 61% higher odds of focal segmental glomerulosclerosis; biallelic APOL1 variants were associated with 25% higher odds of CKD and 84% higher odds of focal segmental glomerulosclerosis. (Funded by the National Human Genome Research Institute and others.).
Collapse
Affiliation(s)
- Rasheed A. Gbadegesin
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina. USA
| | - Ifeoma Ulasi
- Department of Medicine, University of Nigeria, Enugu State, Nigeria
| | - Samuel Ajayi
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Yemi Raji
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Charlotte Osafo
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Adebowale D. Ademola
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adanze Asinobi
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Cheryl A. Winkler
- Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - David Burke
- Departments of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Fatiu Arogundade
- Department of Medicine, Obafemi Awolowo University, Ile- Ife, Nigeria
| | - Ivy Ekem
- Department of Medicine, University of Cape Coast, Cape Coast, Ghana
| | | | - Manmak Mamven
- Department of Medicine, University of Abuja, Abuja, Nigeria
| | - Michael Matekole
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Olukemi Amodu
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Richard Cooper
- Parkinson School of Health Sciences and Public Health, Loyola University, Chicago. USA
| | - Sampson Antwi
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Adebowale A. Adeyemo
- Centre for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Titilayo O. Ilori
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Victoria Adabayeri
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Alexander Nyarko
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana
| | - Toyin Amira
- Department of Medicine, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Adaobi Solarin
- Department of Medicine, College of Medicine, Lagos State University, Lagos, Nigeria
| | - Olugbenga Awobusuyi
- Department of Medicine, College of Medicine, Lagos State University, Lagos, Nigeria
| | - Paul L. Kimmel
- Division of Kidney, Urologic and Digestive Disease, NIDDK, NIH, Bethesda, USA
| | - Frank Chip Brosius
- Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Muhammad Makusidi
- Department of Medicine, Usmanu Danfodiyo University, Sokoto, Sokoto, Nigeria
| | - Uzoma Odenigbo
- Nnamdi Azikiwe University Teaching Hospital, Nnewi, Nigeria
| | - Matthias Kretzler
- Department of Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jeffrey B. Hodgin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Martin R. Pollak
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Vincent Boima
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Barry I. Freedman
- Department of Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Bernard Collins
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Milind Phadnis
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jill Smith
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Celia I. Agwai
- Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ogochukwu Okoye
- Delta State University Teaching Hospital, Oghara, Delta state, Nigeria
| | - Aliyu Abdu
- Aminu Kano Teaching Hospital, Kano, Kano State, Nigeria
| | - Jillian Wilson
- Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Winfred Williams
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Babatunde L. Salako
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Rulan S. Parekh
- Department of Medicine and Pediatrics, Women’s College Hospital, Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Bamidele Tayo
- Parkinson School of Health Sciences and Public Health, Loyola University, Chicago. USA
| | - Dwomoa Adu
- Department of Medicine, University of Ghana Medical School, Accra, Ghana
| | - Akinlolu Ojo
- Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | |
Collapse
|
6
|
Sanna-Cherchi S. APOL1 Kidney Disease Variants - Information from West Africa at Last. N Engl J Med 2025; 392:279-281. [PMID: 39813649 DOI: 10.1056/nejme2415599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Affiliation(s)
- Simone Sanna-Cherchi
- From the Division of Nephrology, Department of Medicine, Columbia University, New York
| |
Collapse
|
7
|
Abu Al Rub F, Elsurer Afsar R, Fleetwood VA, Bastani B, Randall H, Nazzal M, Varma C, Afsar B, Jackson H, Yount S, Wooley C, Light J, Davis V, Caliskan Y, Lentine KL. The Diagnostic Yield of Genomic Sequencing-Based Genetic Kidney Disease Testing in Kidney Transplant Candidates: Experience at an Urban US Transplant Center. Transplantation 2025:00007890-990000000-00982. [PMID: 39819994 DOI: 10.1097/tp.0000000000005288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
BACKGROUND Recent studies suggest that approximately 10% of patients with chronic kidney disease (CKD) have disease-causing genetic variants, an observation relevant to evaluation of kidney transplant candidates. METHODS We retrospectively investigated the diagnostic yield of genetic testing in kidney transplant candidates evaluated at our program (January 1, 2021-December 8, 2022). Inclusion criteria were as follows: first-degree relative(s) with CKD/end-stage kidney disease (ESKD), early-onset CKD, focal segmental glomerulosclerosis, cystic kidney disease, alternative complement pathway-associated diseases, or ESKD of unknown cause. RESULTS One hundred eleven patients underwent genetic kidney disease testing. The most common indication for testing was early-onset CKD (34.2%), followed by a family history of CKD (23.4%), focal segmental glomerulosclerosis (18.0%), cystic kidney disease (9.0%), alternative complement pathway diseases (3.6%), and ESKD of unknown cause (11.7%). Overall diagnostic yield was 46.9% (52/111), and yield was highest among candidates with a family history of CKD (61.5%; 16/26). Among cases with positive testing, the most common diagnostic variant was APOL1, with 2 renal risk variants identified in 57.7% (30/52), while monogenic causes of CKD were identified in 42.3% (22/52). Genetic testing led to further evaluation or to a different diagnosis than the initial clinical diagnosis in 8.1% (9/111) of the cohort. For 24 transplant candidates, their identified diagnostic variants indicated the need for genetic testing of related living donor candidates; of these, 6 living donor candidates were evaluated and underwent testing, of whom donation was excluded in 1 candidate. CONCLUSIONS Pretransplant genetic testing increases understanding of CKD cause, and provides information for living donor evaluation and risk assessment of posttransplant disease recurrence.
Collapse
Affiliation(s)
- Fadee Abu Al Rub
- Saint Louis University Transplant Center, SSM Health Saint Louis University Hospital, St. Louis, MO
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Khan A, Kiryluk K. Polygenic scores and their applications in kidney disease. Nat Rev Nephrol 2025; 21:24-38. [PMID: 39271761 DOI: 10.1038/s41581-024-00886-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/15/2024]
Abstract
Genome-wide association studies (GWAS) have uncovered thousands of risk variants that individually have small effects on the risk of human diseases, including chronic kidney disease, type 2 diabetes, heart diseases and inflammatory disorders, but cumulatively explain a substantial fraction of disease risk, underscoring the complexity and pervasive polygenicity of common disorders. This complexity poses unique challenges to the clinical translation of GWAS findings. Polygenic scores combine small effects of individual GWAS risk variants across the genome to improve personalized risk prediction. Several polygenic scores have now been developed that exhibit sufficiently large effects to be considered clinically actionable. However, their clinical use is limited by their partial transferability across ancestries and a lack of validated models that combine polygenic, monogenic, family history and clinical risk factors. Moreover, prospective studies are still needed to demonstrate the clinical utility and cost-effectiveness of polygenic scores in clinical practice. Here, we discuss evolving methods for developing polygenic scores, best practices for validating and reporting their performance, and the study designs that will empower their clinical implementation. We specifically focus on the polygenic scores relevant to nephrology and other chronic, complex diseases and review their key limitations, necessary refinements and potential clinical applications.
Collapse
Affiliation(s)
- Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
9
|
Zanoni F, Obayemi JE, Gandla D, Castellano G, Keating B. Emerging role of genetics in kidney transplantation. Kidney Int 2024:S0085-2538(24)00913-X. [PMID: 39710162 DOI: 10.1016/j.kint.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 12/24/2024]
Abstract
The advent of more affordable genomic analytical pipelines has facilitated the expansion of genetic studies in kidney transplantation. Advances in genetic sequencing have allowed for a greater understanding of the genetic basis of chronic kidney disease, which has helped to guide transplant management and address issues related to living donation in specific disease settings. Recent efforts have shown significant effects of genetic ancestry and donor APOL1 risk genotypes in determining worse allograft outcomes and increased donation risks. Genetic studies in kidney transplantation outcomes have started to assess the effects of donor and recipient genetics in primary disease recurrence and transplant-related comorbidities, while genome-wide donor-recipient genetic incompatibilities have been shown to represent an important determinant of alloimmunity. Future large-scale comprehensive studies will shed light on the clinical utility of integrative genomics in the kidney transplantation setting.
Collapse
Affiliation(s)
- Francesca Zanoni
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Division of Transplantation, Department of Surgery, New York University Langone Health, Grossman School of Medicine, New York, New York, USA
| | - Joy E Obayemi
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA; Comprehensive Transplant Center, Department of Surgery, Northwestern University, Chicago Illinois, USA
| | - Divya Gandla
- Division of Transplantation, Department of Surgery, New York University Langone Health, Grossman School of Medicine, New York, New York, USA
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Kidney Transplantation, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Brendan Keating
- Institute of Systems Genetics, New York University Langone Health, Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
10
|
Ray PE, Li J, Das J, Xu L, Yu J, Han Z. Pathogenesis of HIV-associated nephropathy in children and adolescents: taking a hard look 40 years later in the era of gene-environment interactions. Am J Physiol Renal Physiol 2024; 327:F1049-F1066. [PMID: 39323389 PMCID: PMC11687833 DOI: 10.1152/ajprenal.00208.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024] Open
Abstract
HIV-associated nephropathy (HIVAN) is a kidney disease that affects mainly people of African ancestry with a high HIV-1 viral load. New antiretroviral therapies (ART) have been highly efficient in preventing and improving the outcome of HIVAN. However, providing chronic ART to children and adolescents living with HIV (CALWH) remains a significant challenge all over the world. More than 2.5 million CALWH, including those living in Sub-Saharan Africa, continue to be at high risk of developing HIVAN. Much of our understanding of the pathogenesis of HIVAN is based on studies conducted in transgenic mice and adults with HIVAN. However, CALWH may experience different health outcomes, risk factors, and susceptibilities to HIVAN in comparison to adults. This article reviews the progress made over the last 40 years in understanding the pathogenesis of HIVAN in CALWH, focusing on how the HIV virus, alongside genetic and environmental factors, contributes to the development of this disease. The landmark discovery that two risk alleles of the apolipoprotein-1 (APOL1) gene play a critical role in HIVAN has significantly advanced our understanding of the disease's pathogenesis. However, we still need to understand why renal inflammation persists despite ART and determine whether the kidney may harbor HIV reservoirs that need to be eliminated to cure HIV permanently. For these reasons, we emphasize reviewing how HIV-1 infects renal cells, affects their growth and regeneration, and discussing how inflammatory cytokines and APOL1 affect the outcome of childhood HIVAN.
Collapse
Affiliation(s)
- Patricio E Ray
- Department of Pediatrics and Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Jinliang Li
- Children's National Hospital, Washington, District of Columbia, United States
| | - Jharna Das
- Children's National Hospital, Washington, District of Columbia, United States
| | - Lian Xu
- Children's National Hospital, Washington, District of Columbia, United States
| | - Jing Yu
- Department of Pediatrics and Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
11
|
Martinelli E, Westland R, Olabisi OA, Sanna-Cherchi S. Prevalence and Impact of APOL1 Kidney Risk Variants in West Africa : Insights for Global Kidney Disease Assessment and Treatment. J Am Soc Nephrol 2024:00001751-990000000-00493. [PMID: 39602366 DOI: 10.1681/asn.0000000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Affiliation(s)
- Elena Martinelli
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
- Unità Operativa Nefrologia, Dipartimento di Medicina e Chirurgia, Azienda Ospedaliero-Universitaria di Parma, Università di Parma, Parma, Italy
| | - Rik Westland
- Department of Pediatric Nephrology, Emma Children's Hospital - Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Opeyemi A Olabisi
- Department of Medicine, Division of Nephrology and Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| |
Collapse
|
12
|
Gonzalez-Vicente A, Crawford DC, Bush WS, Wu Z, Bruggeman LA, Nair V, Eichinger F, Wessely O, Kretzler M, O'Toole JF, Sedor JR. Analysis of Glomerular Transcriptomes from Nephrotic Patients Suggest APOL1 Risk Variants Impact Parietal Epithelial Cells. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.05.24316766. [PMID: 39830251 PMCID: PMC11741451 DOI: 10.1101/2024.11.05.24316766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The disproportionate risk for idiopathic proteinuric podocytopathies in Black people is explained, in part, by the presence of two risk alleles (G1 or G2) in the APOL1 gene. The pathogenic mechanisms responsible for this genetic association remain incompletely understood. We analyzed glomerular RNASeq transcriptomes from patients with idiopathic nephrotic syndrome of which 72 had inferred African ancestry (AA) and 152 did not (noAA). Using gene coexpression networks we found a significant association between APOL1 risk allele number and the coexpression metamodule 2 (MM2), even after adjustment for eGFR and proteinuria at biopsy. Unadjusted Kaplan-Meier curves showed that unlike noAA, AA with the highest tertile of MM2 gene activation scores were less likely to achieve complete remission (p≤0.014). Characteristic direction (ChDir) identified a signature of 1481 genes, which separated patients with APOL1 risk alleles from those homozygous for reference APOL1 . Only in AA, the tertile with the highest activation scores of these 1481 genes was less likely to achieve complete remission (p≤0.022) and showed a trend to faster progression to the composite event of kidney failure or loss of 40% eGFR (p≤0.099). The MM2 and ChDir genes significantly overlapped and were both enriched for Epithelial Mesenchymal Transition and inflammation terms. Finally, MM2 significantly overlapped with a parietal epithelial cell (PEC)-identity gene signature but not with a podocyte identity signature. Podocytes expressing variant APOL1s may generate inflammatory signals that activate PECs by paracrine mechanisms contributing to APOL1 nephropathy.
Collapse
|
13
|
Weiss R, Milo Rasouly H, Marasa M, Fernandez H, Lin F, Sabatello M. Nephrologists' Views on a Workflow for Returning Genetic Results to Research Participants. Kidney Int Rep 2024; 9:3278-3289. [PMID: 39534211 PMCID: PMC11551134 DOI: 10.1016/j.ekir.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Returning research-based genetic results (gRoR) to participants in nephrology research can improve care; however, the practice raises implementational questions and no established guidelines for this process currently exist. Nephrologists' views on this issue can inform the process but are understudied. Methods We developed a conceptual workflow for gRoR from literature and experience, covering aspects such as which results to return, how, and by whom. We surveyed US nephrologists to gauge their views on the workflow and anticipated barriers and collected participants' demographics, including professional backgrounds. Results A total of 201 adult and pediatric nephrologists completed the survey. Most of them agreed that all diagnostic kidney-related results (93%), secondary findings (80%), and kidney-related risk variants (83%) should be returned. No significant differences were found between adult and pediatric nephrologists' responses, except that 48% of adult nephrologists versus 26% of pediatric nephrologists supported returning polygenic risk scores (PRS) (P < 0.01). Seventy-nine percent wanted to know about research results before clinical confirmation. Most of them (63%) believed a genetic counselor should return clinically confirmed results. Key barriers included the cost of clinical validation (77%) and the unavailability of genetic counseling services (63%). Facilitators included educational resources on genetic kidney diseases (91%), a referral list of experts (89%), and clear clinical care guidelines (89%). We discuss findings' implications and provide "points to consider." Conclusion There is significant interest in gRoR among nephrologists; however, logistical and economic concerns need addressing. Identified facilitators can help large nephrology studies planning to return genetic results to participants.
Collapse
Affiliation(s)
- Robyn Weiss
- Department of Obstetrics and Gynecology, Maimonides Medical Center, Brooklyn, New York, New York, USA
- Sarah Lawrence College Joan H. Marks Graduate Program in Human Genetics, Bronxville, New York, USA
| | - Hila Milo Rasouly
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York, USA
| | - Maddalena Marasa
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York, USA
| | - Hilda Fernandez
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York, USA
| | - Fangming Lin
- Division of Pediatric Nephrology, Department of Pediatrics, Columbia University, New York, New York, USA
| | - Maya Sabatello
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York, USA
- Division of Ethics, Department of Medical Humanities and Ethics, Columbia University, New York, New York, USA
| |
Collapse
|
14
|
Mann N, Sun H, Majmundar AJ. Mechanisms of podocyte injury in genetic kidney disease. Pediatr Nephrol 2024:10.1007/s00467-024-06551-x. [PMID: 39485497 DOI: 10.1007/s00467-024-06551-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024]
Abstract
Glomerular diseases are a leading cause of chronic kidney disease worldwide. Both acquired and hereditary glomerulopathies frequently share a common final disease mechanism: disruption of the glomerular filtration barrier, podocyte injury, and ultimately podocyte death and detachment. Over 70 monogenic causes of proteinuric kidney disease have been identified, and most of these genes are highly expressed in podocytes, regulating key processes such as maintenance of the slit diaphragm, regulation of actin cytoskeleton remodeling, and modulation of downstream transcriptional pathways. Collectively, these are increasingly being referred to as hereditary "podocytopathies," in which podocyte injury is the central feature driving proteinuria and kidney dysfunction. In this review, we provide an overview of the monogenic podocytopathies and discuss the molecular mechanisms by which single-gene defects lead to podocyte injury and ultimately glomerulosclerosis. We review how advances in genomic technology and a better understanding of the cell biological basis of disease have led to the development of more targeted and personalized therapeutic strategies, including an overview of small molecule and gene therapy approaches.
Collapse
Affiliation(s)
- Nina Mann
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Hua Sun
- Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amar J Majmundar
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
15
|
Pays E. Apolipoprotein-L1 (APOL1): From Sleeping Sickness to Kidney Disease. Cells 2024; 13:1738. [PMID: 39451256 PMCID: PMC11506758 DOI: 10.3390/cells13201738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Apolipoprotein-L1 (APOL1) is a membrane-interacting protein induced by inflammation, which confers human resistance to infection by African trypanosomes. APOL1 kills Trypanosoma brucei through induction of apoptotic-like parasite death, but two T. brucei clones acquired resistance to APOL1, allowing them to cause sleeping sickness. An APOL1 C-terminal sequence alteration, such as occurs in natural West African variants G1 and G2, restored human resistance to these clones. However, APOL1 unfolding induced by G1 or G2 mutations enhances protein hydrophobicity, resulting in kidney podocyte dysfunctions affecting renal filtration. The mechanism involved in these dysfunctions is debated. The ability of APOL1 to generate ion pores in trypanosome intracellular membranes or in synthetic membranes was provided as an explanation. However, transmembrane insertion of APOL1 strictly depends on acidic conditions, and podocyte cytopathology mainly results from secreted APOL1 activity on the plasma membrane, which occurs under non-acidic conditions. In this review, I argue that besides inactivation of APOL3 functions in membrane dynamics (fission and fusion), APOL1 variants induce inflammation-linked podocyte toxicity not through pore formation, but through plasma membrane disturbance resulting from increased interaction with cholesterol, which enhances cation channels activity. A natural mutation in the membrane-interacting domain (N264K) abrogates variant APOL1 toxicity at the expense of slightly increased sensitivity to trypanosomes, further illustrating the continuous mutual adaptation between host and parasite.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, Institut de Biologie et de Médecine Moléculaires (IBMM), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| |
Collapse
|
16
|
Olabisi OA, Barrett NJ, Lucas A, Smith M, Bethea K, Soldano K, Croall S, Sadeghpour A, Chakraborty H, Wolf M. Design and Rationale of the Phase 2 Baricitinib Study in Apolipoprotein L1-Mediated Kidney Disease (JUSTICE). Kidney Int Rep 2024; 9:2677-2684. [PMID: 39291185 PMCID: PMC11403079 DOI: 10.1016/j.ekir.2024.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction Individuals of recent West African ancestry develop focal segmental glomerulosclerosis (FSGS) and hypertension-attributed end-stage kidney disease (HTN-ESKD) at 4 times the rate of White Americans. Two protein-coding variants of the Apolipoprotein L1 (APOL1) gene, G1 and G2, explain 50% to 70% of the excess risk of HTN-ESKD and FSGS among this group. Increased expression of G1 and G2 in the kidney, mediated by Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling, drive pathogenesis of these kidney diseases. Baricitinib is an orally active inhibitor of JAK1/2 that blocks APOL1 synthesis. The Janus kinase-STAT Inhibition to Reduce APOL1-Associated Kidney Disease (JUSTICE) trial is evaluating the antiproteinuric efficacy and safety of baricitinib in patients with APOL1-associated FSGS and HTN-attributed chronic kidney disease (HTN-CKD). Methods JUSTICE is a single-center, randomized, double-blind, placebo-controlled, pilot phase 2 trial of baricitinib in patients with proteinuria, APOL1-associated FSGS or APOL1-associated HTN-CKD without diabetes. A total of 75 African American patients with APOL1-associated CKD, including 25 with FSGS and 50 with HTN-CKD, aged 18 to 70 years will be randomized 2:1 to daily treatment with baricitinib or placebo, respectively. Results The primary efficacy end point will be percent change in urine albumin-to-creatinine ratio (UACR) from baseline to end of month 6. The primary safety end point will be incidence of clinically significant decreases in hemoglobin of ≥ 1g/dl. Conclusion The phase 2 JUSTICE study will characterize the antiproteinuric efficacy and safety of JAK1/2 inhibition with baricitinib in patients with APOL1-associated FSGS and APOL1-associated HTN-CKD.
Collapse
Affiliation(s)
- Opeyemi A Olabisi
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Nadine J Barrett
- Atrium Health/Wake Forest Comprehensive Cancer Center and Maya Angelo Center for Health Equity, Wake Forest School of Medicine, Wake Forest, North Carolina, USA
- Department of Social Science and Health Policy, Division of Population Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Maya Angelo Center for Health Equity, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anika Lucas
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Maurice Smith
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kenisha Bethea
- Duke Clinical and Translational Science Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Karen Soldano
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Stephanie Croall
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Azita Sadeghpour
- Duke Precision Medicine Program, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Myles Wolf
- Division of Nephrology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
17
|
Nystrom SE, Soldano KL, Rockett M, Datta S, Li G, Silas D, Garrett ME, Ashley-Koch AE, Olabisi OA. APOL1 High-Risk Genotype is Not Associated With New or Worsening of Proteinuria or Kidney Function Decline Following COVID-19 Vaccination. Kidney Int Rep 2024; 9:2657-2666. [PMID: 39291186 PMCID: PMC11403097 DOI: 10.1016/j.ekir.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction SARS-CoV-2 infection increases systemic inflammatory cytokines which act as a second-hit driver of Apolipoprotein L1 (APOL1)-mediated collapsing glomerulopathy. SARS-CoV-2 vaccination also increases cytokines. Recent reports of new glomerular disease in individuals with APOL1 high-risk genotype (HRG) following SARS-CoV-2 vaccination raised the concern SARS-CoV-2 vaccination may also act as a second-hit driver of APOL1-mediated glomerulopathy. Methods We screened 1507 adults in the Duke's Measurement to Understand Reclassification of Disease of Cabarrus and Kannapolis (MURDOCK) registry and enrolled 105 eligible participants with available SARS-CoV-2 vaccination data, prevaccination and postvaccination serum creatinine, and urine protein measurements. Paired data were stratified by number of APOL1 risk alleles (RAs) and compared within groups using Wilcoxon signed rank test and across groups by analysis of variance. Results Among 105 participants, 30 (28.6%) had 2, 39 (37.1%) had 1, and 36 (34.3%) had 0 APOL1 RA. Most of the participants (94%) received at least 2 doses of vaccine. Most (98%) received the BNT162B2 (Pfizer) or mRNA-1273 (Moderna) vaccine. On average, the prevaccine and postvaccine laboratory samples were drawn 648 days apart. There were no detectable differences between pre- and post-serum creatinine or pre- and post-urine albumin creatinine ratio irrespective of the participants' APOL1 genotype. Finally, most participants with APOL1 RA had the most common haplotype (E150, I228, and K255) and lacked the recently described protective N264K haplotype. Conclusion In this observational study, APOL1 HRG is not associated with new or worsening of proteinuria or decline in kidney function following SARS-CoV-2 vaccination. Validation of this result in larger cohorts would further support the renal safety of SARS-CoV-2 vaccine in individuals with APOL1 HRG.
Collapse
Affiliation(s)
- Sarah E Nystrom
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Karen L Soldano
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Micki Rockett
- Duke Clinical and Translational Science Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Somenath Datta
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Guojie Li
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Daniel Silas
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Melanie E Garrett
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Allison E Ashley-Koch
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Opeyemi A Olabisi
- Division of Nephrology, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
18
|
Madhavan SM, Schlöndorff JS. Variant upon variant: kidney-disease risk associated with APOL1 G2 genetic variants is abrogated by the APOL1 p.N264K variant. Kidney Int 2024; 106:345-348. [PMID: 38750901 DOI: 10.1016/j.kint.2024.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/12/2024] [Indexed: 05/25/2024]
Affiliation(s)
- Sethu M Madhavan
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Johannes S Schlöndorff
- Division of Nephrology, Department of Medicine, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
19
|
Gulati A, Moxey-Mims M. Defining Risk in APOL1-Associated Kidney Disease: The Story is Evolving! Am J Kidney Dis 2024; 84:388-391. [PMID: 38648881 DOI: 10.1053/j.ajkd.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024]
Affiliation(s)
- Ashima Gulati
- Division of Pediatric Nephrology, Children's National Hospital, Washington, DC; Department of Pediatrics, The George Washington University School of Medicine, Washington, DC
| | - Marva Moxey-Mims
- Division of Pediatric Nephrology, Children's National Hospital, Washington, DC; Department of Pediatrics, The George Washington University School of Medicine, Washington, DC.
| |
Collapse
|
20
|
Vivante A. Genetics of Chronic Kidney Disease. N Engl J Med 2024; 391:627-639. [PMID: 39141855 DOI: 10.1056/nejmra2308577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Affiliation(s)
- Asaf Vivante
- From the Department of Pediatrics and the Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, and the Nephro-Genetics Clinic and Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel Hashomer, and the Faculty of Medicine, Tel Aviv University, Tel Aviv - all in Israel
| |
Collapse
|
21
|
Caparali EB, De Gregorio V, Barua M. Genetic Causes of Nephrotic Syndrome and Focal and Segmental Glomerulosclerosis. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:309-316. [PMID: 39084756 DOI: 10.1053/j.akdh.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 08/02/2024]
Abstract
The field of nephrology has a long-standing interest in deciphering the genetic basis of nephrotic syndrome (NS), motivated by the mechanistic insights it provides in chronic kidney disease. The initial era of genetic studies solidified NS and the focal segmental glomerulosclerosis lesion as podocyte disorders. The likelihood of identifying a single gene (called monogenic) cause is higher if certain factors are present such as positive family history. Obtaining a monogenic diagnosis enables reproductive counseling and screening of family members. Now, with a new era of genomic studies facilitated by technological advances and the emergence of large genetically characterized cohorts, more insights are apparent. This includes the phenotypic breadth associated with disease genes, as evidenced in Alport syndrome and congenital NS of the Finnish type. Moreover, the underlying genetic architecture is more complex than previously appreciated, as shown by genome-wide association studies, suggesting that variants in multiple genes collectively influence risk. Achieving molecularly informed diagnoses also holds substantial potential for personalizing medicine, including the development of targeted therapeutics. Illustrative examples include coenzyme Q10 for ADCK4-associated NS and inaxaplin, a small molecule that inhibits apolipoprotein L1 channel activity, though larger studies are required to confirm benefit.
Collapse
Affiliation(s)
- Emine Bilge Caparali
- Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Vanessa De Gregorio
- Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Moumita Barua
- Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Tabachnikov O, Skorecki K, Kruzel-Davila E. APOL1 nephropathy - a population genetics success story. Curr Opin Nephrol Hypertens 2024; 33:447-455. [PMID: 38415700 PMCID: PMC11139250 DOI: 10.1097/mnh.0000000000000977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
PURPOSE OF REVIEW More than a decade ago, apolipoprotein L1 ( APOL1 ) risk alleles designated G1 and G2, were discovered to be causally associated with markedly increased risk for progressive kidney disease in individuals of recent African ancestry. Gratifying progress has been made during the intervening years, extending to the development and clinical testing of genomically precise small molecule therapy accompanied by emergence of RNA medicine platforms and clinical testing within just over a decade. RECENT FINDINGS Given the plethora of excellent prior review articles, we will focus on new findings regarding unresolved questions relating mechanism of cell injury with mode of inheritance, regulation and modulation of APOL1 activity, modifiers and triggers for APOL1 kidney risk penetrance, the pleiotropic spectrum of APOL1 related disease beyond the kidney - all within the context of relevance to therapeutic advances. SUMMARY Notwithstanding remaining controversies and uncertainties, promising genomically precise therapies targeted at APOL1 mRNA using antisense oligonucleotides (ASO), inhibitors of APOL1 expression, and small molecules that specifically bind and inhibit APOL1 cation flux are emerging, many already at the clinical trial stage. These therapies hold great promise for mitigating APOL1 kidney injury and possibly other systemic phenotypes as well. A challenge will be to develop guidelines for appropriate use in susceptible individuals who will derive the greatest benefit.
Collapse
Affiliation(s)
- Orly Tabachnikov
- Department of Nephrology, Rambam Healthcare Campus, Haifa, Israel
| | - Karl Skorecki
- Department of Nephrology, Rambam Healthcare Campus, Haifa, Israel
- Departments of Genetics and Developmental Biology and Rappaport Faculty of Medicine and Research Institute, Technion—Israel Institute of Technology, Haifa, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Etty Kruzel-Davila
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Department of Nephrology, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
23
|
Narjoz C, Vinh-Hoang-Lan Julie Tran, Rabant M, Karras A, Pallet N. Diagnostic Yield of APOL1 p.N264K Variant Screening in Daily Practice. Kidney Int Rep 2024; 9:1916-1918. [PMID: 38899219 PMCID: PMC11184237 DOI: 10.1016/j.ekir.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 06/21/2024] Open
Affiliation(s)
- Céline Narjoz
- Department of Clinical Chemistry, Assistance Publique Hôpitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Vinh-Hoang-Lan Julie Tran
- Department of Clinical Chemistry, Assistance Publique Hôpitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Marion Rabant
- Department of Pathology, Assistance Publique Hôpitaux de Paris, Necker Hospital, Paris, France
- University Paris Cité, Paris, France
| | - Alexandre Karras
- University Paris Cité, Paris, France
- Department of Nephrology, Assistance Publique Hôpitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Nicolas Pallet
- Department of Clinical Chemistry, Assistance Publique Hôpitaux de Paris, Georges Pompidou European Hospital, Paris, France
- Department of Nephrology, Assistance Publique Hôpitaux de Paris, Georges Pompidou European Hospital, Paris, France
| |
Collapse
|
24
|
Palmer ND, Freedman BI. APOL1-mediated and Mendelian forms of "heretofore" idiopathic collapsing glomerulopathy: lessons from Brazil. Kidney Int 2024; 105:437-439. [PMID: 38388143 DOI: 10.1016/j.kint.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 02/24/2024]
Abstract
APOL1-mediated kidney diseases have forever changed nephrology and kidney transplantation. Neves et al. extend this field with analyses in admixed Brazilians with the most severe type of APOL1-mediated kidney disease, idiopathic collapsing glomerulopathy. Causative gene variants were detected in 58.6% of patients; 80.5% had APOL1 high-risk genotypes, and 19.5% had causative Mendelian variants. Their work identifies the cause of previous idiopathic collapsing glomerulopathy and provides opportunities to identify novel modifiers in severe APOL1-mediated kidney diseases that are relevant beyond Brazil.
Collapse
Affiliation(s)
- Nicholette D Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Barry I Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
25
|
Datta S, Antonio BM, Zahler NH, Theile JW, Krafte D, Zhang H, Rosenberg PB, Chaves AB, Muoio DM, Zhang G, Silas D, Li G, Soldano K, Nystrom S, Ferreira D, Miller SE, Bain JR, Muehlbauer MJ, Ilkayeva O, Becker TC, Hohmeier HE, Newgard CB, Olabisi OA. APOL1-mediated monovalent cation transport contributes to APOL1-mediated podocytopathy in kidney disease. J Clin Invest 2024; 134:e172262. [PMID: 38227370 PMCID: PMC10904047 DOI: 10.1172/jci172262] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024] Open
Abstract
Two coding variants of apolipoprotein L1 (APOL1), called G1 and G2, explain much of the excess risk of kidney disease in African Americans. While various cytotoxic phenotypes have been reported in experimental models, the proximal mechanism by which G1 and G2 cause kidney disease is poorly understood. Here, we leveraged 3 experimental models and a recently reported small molecule blocker of APOL1 protein, VX-147, to identify the upstream mechanism of G1-induced cytotoxicity. In HEK293 cells, we demonstrated that G1-mediated Na+ import/K+ efflux triggered activation of GPCR/IP3-mediated calcium release from the ER, impaired mitochondrial ATP production, and impaired translation, which were all reversed by VX-147. In human urine-derived podocyte-like epithelial cells (HUPECs), we demonstrated that G1 caused cytotoxicity that was again reversible by VX-147. Finally, in podocytes isolated from APOL1 G1 transgenic mice, we showed that IFN-γ-mediated induction of G1 caused K+ efflux, activation of GPCR/IP3 signaling, and inhibition of translation, podocyte injury, and proteinuria, all reversed by VX-147. Together, these results establish APOL1-mediated Na+/K+ transport as the proximal driver of APOL1-mediated kidney disease.
Collapse
Affiliation(s)
- Somenath Datta
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Duke University School of Medicine, Department of Medicine, Division of Nephrology, Durham, North Carolina, USA
| | | | | | | | | | - Hengtao Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Paul B. Rosenberg
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alec B. Chaves
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Deborah M. Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Guofang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| | - Daniel Silas
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Duke University School of Medicine, Department of Medicine, Division of Nephrology, Durham, North Carolina, USA
| | - Guojie Li
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Duke University School of Medicine, Department of Medicine, Division of Nephrology, Durham, North Carolina, USA
| | - Karen Soldano
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Duke University School of Medicine, Department of Medicine, Division of Nephrology, Durham, North Carolina, USA
| | - Sarah Nystrom
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Duke University School of Medicine, Department of Medicine, Division of Nephrology, Durham, North Carolina, USA
| | - Davis Ferreira
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sara E. Miller
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - James R. Bain
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| | - Michael J. Muehlbauer
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| | - Thomas C. Becker
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hans-Ewald Hohmeier
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
| | - Christopher B. Newgard
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Opeyemi A. Olabisi
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
- Duke University School of Medicine, Department of Medicine, Division of Nephrology, Durham, North Carolina, USA
| |
Collapse
|
26
|
Gbadegesin R, Martinelli E, Gupta Y, Friedman DJ, Sampson MG, Pollak MR, Sanna-Cherchi S. APOL1 Genotyping Is Incomplete without Testing for the Protective M1 Modifier p.N264K Variant. GLOMERULAR DISEASES 2024; 4:43-48. [PMID: 38495868 PMCID: PMC10942791 DOI: 10.1159/000537948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/16/2024] [Indexed: 03/19/2024]
Affiliation(s)
- Rasheed Gbadegesin
- Division of Nephrology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Elena Martinelli
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Unità Operativa Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
| | - Yask Gupta
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Institute for Inflammation Medicine, University of Lübeck, Lübeck, Germany
| | - David J Friedman
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Matthew G Sampson
- Harvard Medical School, Boston, MA, USA
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative and Medical and Population Genetics Program, Broad Institute, Boston, MA, USA
- Division of Nephrology, Brigham and Women's Hospital, Boston, MA, USA
| | - Martin R Pollak
- Nephrology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Simone Sanna-Cherchi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|