101
|
Doumatey AP, Bentley AR, Akinyemi R, Olanrewaju TO, Adeyemo A, Rotimi C. Genes, environment, and African ancestry in cardiometabolic disorders. Trends Endocrinol Metab 2023; 34:601-621. [PMID: 37598069 PMCID: PMC10548552 DOI: 10.1016/j.tem.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
The past two decades have been characterized by a substantial global increase in cardiometabolic diseases, but the prevalence and incidence of these diseases and related traits differ across populations. African ancestry populations are among the most affected yet least included in research. Populations of African descent manifest significant genetic and environmental diversity and this under-representation is a missed opportunity for discovery and could exacerbate existing health disparities and curtail equitable implementation of precision medicine. Here, we discuss cardiometabolic diseases and traits in the context of African descent populations, including both genetic and environmental contributors and emphasizing novel discoveries. We also review new initiatives to include more individuals of African descent in genomics to address current gaps in the field.
Collapse
Affiliation(s)
- Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rufus Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training and Centre for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria; Department of Neurology, University College Hospital, Ibadan, Nigeria
| | - Timothy O Olanrewaju
- Division of Nephrology, Department of Medicine, University of Ilorin & University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Charles Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
102
|
Qi Z, Yuan S, Wei J, Xia S, Huang Y, Chen X, Han Y, Li Z, Xiao Y, Peng F, Fu X, Sun L, Liu H, Zhu X. Clinical and pathological features of omicron variant of SARS-CoV-2-associated kidney injury. J Med Virol 2023; 95:e29196. [PMID: 37881096 DOI: 10.1002/jmv.29196] [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: 06/21/2023] [Revised: 10/07/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023]
Abstract
Kidney injury is common in patients with Coronavirus Disease-19 (COVID-19), which is related to poor prognosis. We aim to summarize the clinical features, athological types, and prognosis of COVID-19 associated kidney injury caused by the Omicron strain. In this study, 46 patients with Omicron-associated kidney injury were included, 38 of whom performed renal biopsy. Patients were divided into two groups: group A for patients with onset of kidney injury after SARS-CoV-2 infection; group B for patients with pre-existing kidney disease who experienced aggravation of renal insufficiency after SARS-CoV-2 infection. The clinical, pathological, and prognostic characteristics of the patients were observed. Acute kidney injury (AKI) (35%) was the most common clinical manifestation in group A. Patients in group B mainly presented with chronic kidney disease (CKD) (55%) and nephrotic syndrome (NS) (40%). The pathological type was mainly IgA nephropathy (IgAN) (39% in group A and 45% in group B). Among all of them, one case presenting with thrombotic microangiopathy had worse kidney function at biopsy time. Mean serum C3 levels were 1.2 ± 0.5 and 1.0 ± 0.2 g/L in group A and group B, respectively. In renal tissues, C3 deposits were observed in 71.1% of patients. 11.8% (n = 2) patients experienced deterioration of renal function after treatment, but no patients developed to end-stage renal disease. In our single-center study in China, the main clinical manifestations were AKI, CKD, and NS, while the main pathological type was IgAN. Compared with previous strains of SARS-CoV-2, patients with the Omicron infection had a favorable prognosis.
Collapse
Affiliation(s)
- Zhiwen Qi
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuguang Yuan
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jinying Wei
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shiyu Xia
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yao Huang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaojun Chen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yachun Han
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zheng Li
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yang Xiao
- Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fenghua Peng
- Department of Kidney Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiao Fu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lin Sun
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hong Liu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xuejing Zhu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
103
|
Hampton G, Kim J, Edwards TL, Hellwege JN, Velez Edwards DR. Uterine leiomyomata and keloids fibrosis origins: a mini-review of fibroproliferative diseases. Am J Physiol Cell Physiol 2023; 325:C817-C822. [PMID: 37642233 PMCID: PMC10635651 DOI: 10.1152/ajpcell.00181.2023] [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: 05/02/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
Diseases such as uterine leiomyomata (fibroids and benign tumors of the uterus) and keloids (raised scars) may share common etiology. Fibroids and keloids can co-occur in individuals, and both are highly heritable, suggesting they may share common genetic risk factors. Fibroproliferative diseases are common and characterized by scarring and overgrowth of connective tissue, impacting multiple organ systems. These conditions both have racial disparities in prevalence, with the highest prevalence observed among individuals of African ancestry. Several fibroproliferative diseases are more severe and common in populations of sub-Saharan Africa. This mini-review aims to provide a broad overview of the current knowledge of the evolutionary origins and causes of fibroproliferative diseases. We also discuss current hypotheses proposing that the increased prevalence of these diseases in African-derived populations is due to the selection for profibrotic alleles that are protective against helminth infections and provide examples from knowledge of uterine fibroid and keloid research.
Collapse
Affiliation(s)
- Gabrielle Hampton
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jeewoo Kim
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Todd L Edwards
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jacklyn N Hellwege
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
104
|
Naas S, Schiffer M, Schödel J. Hypoxia and renal fibrosis. Am J Physiol Cell Physiol 2023; 325:C999-C1016. [PMID: 37661918 DOI: 10.1152/ajpcell.00201.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Renal fibrosis is the final stage of most progressive kidney diseases. Chronic kidney disease (CKD) is associated with high comorbidity and mortality. Thus, preventing fibrosis and thereby preserving kidney function increases the quality of life and prolongs the survival of patients with CKD. Many processes such as inflammation or metabolic stress modulate the progression of kidney fibrosis. Hypoxia has also been implicated in the pathogenesis of renal fibrosis, and oxygen sensing in the kidney is of outstanding importance for the body. The dysregulation of oxygen sensing in the diseased kidney is best exemplified by the loss of stimulation of erythropoietin production from interstitial cells in the fibrotic kidney despite anemia. Furthermore, hypoxia is present in acute or chronic kidney diseases and may affect all cell types present in the kidney including tubular and glomerular cells as well as resident immune cells. Pro- and antifibrotic effects of the transcription factors hypoxia-inducible factors 1 and 2 have been described in a plethora of animal models of acute and chronic kidney diseases, but recent advances in sequencing technologies now allow for novel and deeper insights into the role of hypoxia and its cell type-specific effects on the progression of renal fibrosis, especially in humans. Here, we review existing literature on how hypoxia impacts the development and progression of renal fibrosis.
Collapse
Affiliation(s)
- Stephanie Naas
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
105
|
Liu M, Ning Y, Teixayavong S, Mertens M, Xu J, Ting DSW, Cheng LTE, Ong JCL, Teo ZL, Tan TF, RaviChandran N, Wang F, Celi LA, Ong MEH, Liu N. A translational perspective towards clinical AI fairness. NPJ Digit Med 2023; 6:172. [PMID: 37709945 PMCID: PMC10502051 DOI: 10.1038/s41746-023-00918-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023] Open
Abstract
Artificial intelligence (AI) has demonstrated the ability to extract insights from data, but the fairness of such data-driven insights remains a concern in high-stakes fields. Despite extensive developments, issues of AI fairness in clinical contexts have not been adequately addressed. A fair model is normally expected to perform equally across subgroups defined by sensitive variables (e.g., age, gender/sex, race/ethnicity, socio-economic status, etc.). Various fairness measurements have been developed to detect differences between subgroups as evidence of bias, and bias mitigation methods are designed to reduce the differences detected. This perspective of fairness, however, is misaligned with some key considerations in clinical contexts. The set of sensitive variables used in healthcare applications must be carefully examined for relevance and justified by clear clinical motivations. In addition, clinical AI fairness should closely investigate the ethical implications of fairness measurements (e.g., potential conflicts between group- and individual-level fairness) to select suitable and objective metrics. Generally defining AI fairness as "equality" is not necessarily reasonable in clinical settings, as differences may have clinical justifications and do not indicate biases. Instead, "equity" would be an appropriate objective of clinical AI fairness. Moreover, clinical feedback is essential to developing fair and well-performing AI models, and efforts should be made to actively involve clinicians in the process. The adaptation of AI fairness towards healthcare is not self-evident due to misalignments between technical developments and clinical considerations. Multidisciplinary collaboration between AI researchers, clinicians, and ethicists is necessary to bridge the gap and translate AI fairness into real-life benefits.
Collapse
Affiliation(s)
- Mingxuan Liu
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Yilin Ning
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | | | - Mayli Mertens
- Centre for Ethics, Department of Philosophy, University of Antwerp, Antwerp, Belgium
- Antwerp Center on Responsible AI, University of Antwerp, Antwerp, Belgium
| | - Jie Xu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Daniel Shu Wei Ting
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- SingHealth AI Office, Singapore Health Services, Singapore, Singapore
| | - Lionel Tim-Ee Cheng
- Department of Diagnostic Radiology, Singapore General Hospital, Singapore, Singapore
| | | | - Zhen Ling Teo
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Ting Fang Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | | | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Leo Anthony Celi
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marcus Eng Hock Ong
- Programme in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
- Department of Emergency Medicine, Singapore General Hospital, Singapore, Singapore
| | - Nan Liu
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore.
- SingHealth AI Office, Singapore Health Services, Singapore, Singapore.
- Programme in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore.
- Institute of Data Science, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
106
|
Jiang L, Liu G, Oeser A, Ihegword A, Dickson AL, Daniel LL, Hung AM, Cox NJ, Chung CP, Wei WQ, Stein CM, Feng Q. Association between APOL1 risk variants and the occurrence of sepsis in Black patients hospitalized with infections: a retrospective cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.27.23284540. [PMID: 36747677 PMCID: PMC9901067 DOI: 10.1101/2023.01.27.23284540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Background Two risk variants in the apolipoprotein L1 gene ( APOL1 ) have been associated with increased susceptibility to sepsis in Black patients. However, it remains unclear whether APOL1 high-risk genotypes are associated with occurrence of either sepsis or sepsis-related phenotypes in patients hospitalized with infections, independent of their association with pre-existing severe renal disease. Methods A retrospective cohort study of 2,242 Black patients hospitalized with infections. We assessed whether carriage of APOL1 high-risk genotypes was associated with the risk of sepsis and sepsis-related phenotypes in patients hospitalized with infections. The primary outcome was sepsis; secondary outcomes were short-term mortality and organ failure related to sepsis. Results Of 2,242 Black patients hospitalized with infections, 565 developed sepsis. Patients with high-risk APOL1 genotypes had a significantly increased risk of sepsis (odds ratio [OR]=1.29 [95% CI, 1.00-1.67; p=0.047]); however, this association was not significant after adjustment for pre-existing severe renal disease (OR=1.14 [95% CI, 0.88-1.48; p=0.33]), nor after exclusion of those patients with pre-existing severe renal disease (OR=0.99 [95% CI, 0.70-1.39; p=0.95]. APOL1 high-risk genotypes were significantly associated with the renal dysfunction component of the Sepsis-3 criteria (OR=1.64 [95% CI, 1.21-2.22; p=0.001], but not with other sepsis-related organ dysfunction or short-term mortality. The association between high-risk APOL1 genotypes and sepsis-related renal dysfunction was markedly attenuated by adjusting for pre-existing severe renal disease (OR=1.36 [95% CI, 1.00-1.86; p=0.05]) and was nullified after exclusion of patients with pre-existing severe renal disease (OR=1.16 [95% CI, 0.74-1.81; p=0.52]). Conclusion APOL1 high-risk genotypes were associated with an increased risk of sepsis; however, this increased risk was attributable predominantly to pre-existing severe renal disease. Funding This study was supported by R01GM120523 (Q.F.), R01HL163854 (Q.F.), R35GM131770 (C.M.S.), HL133786 (W.Q.W.), and Vanderbilt Faculty Research Scholar Fund (Q.F.). The dataset(s) used for the analyses described were obtained from Vanderbilt University Medical Center's BioVU which is supported by institutional funding, the 1S10RR025141-01 instrumentation award, and by the CTSA grant UL1TR0004from NCATS/NIH. Additional funding provided by the NIH through grants P50GM115305 and U19HL065962. The authors wish to acknowledge the expert technical support of the VANTAGE and VANGARD core facilities, supported in part by the Vanderbilt-Ingram Cancer Center (P30 CA068485) and Vanderbilt Vision Center (P30 EY08126).The funders had no role in design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Collapse
|
107
|
Garrett ME, Soldano KL, Erwin KN, Zhang Y, Gordeuk VR, Gladwin MT, Telen MJ, Ashley-Koch AE. Genome-wide meta-analysis identifies new candidate genes for sickle cell disease nephropathy. Blood Adv 2023; 7:4782-4793. [PMID: 36399516 PMCID: PMC10469559 DOI: 10.1182/bloodadvances.2022007451] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 10/11/2022] [Accepted: 10/29/2022] [Indexed: 11/19/2022] Open
Abstract
Sickle cell disease nephropathy (SCDN), a common SCD complication, is strongly associated with mortality. Polygenic risk scores calculated from recent transethnic meta-analyses of urinary albumin-to-creatinine ratio and estimated glomerular filtration rate (eGFR) trended toward association with proteinuria and eGFR in SCD but the model fit was poor (R2 < 0.01), suggesting that there are likely unique genetic risk factors for SCDN. Therefore, we performed genome-wide association studies (GWAS) for 2 critical manifestations of SCDN, proteinuria and decreased eGFR, in 2 well-characterized adult SCD cohorts, representing, to the best of our knowledge, the largest SCDN sample to date. Meta-analysis identified 6 genome-wide significant associations (false discovery rate, q ≤ 0.05): 3 for proteinuria (CRYL1, VWF, and ADAMTS7) and 3 for eGFR (LRP1B, linc02288, and FPGT-TNNI3K/TNNI3K). These associations are independent of APOL1 risk and represent novel SCDN loci, many with evidence for regulatory function. Moreover, GWAS SNPs in CRYL1, VWF, ADAMTS7, and linc02288 are associated with gene expression in kidney and pathways important to both renal function and SCD biology, supporting the hypothesis that SCDN pathophysiology is distinct from other forms of kidney disease. Together, these findings provide new targets for functional follow-up that could be tested prospectively and potentially used to identify patients with SCD who are at risk, before onset of kidney dysfunction.
Collapse
Affiliation(s)
- Melanie E. Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC
| | - Karen L. Soldano
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC
| | - Kyle N. Erwin
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Mark T. Gladwin
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Marilyn J. Telen
- Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC
| | | |
Collapse
|
108
|
Nakagawa N, Kimura T, Sakate R, Wada T, Furuichi K, Okada H, Isaka Y, Narita I. Demographics and treatment of patients with primary nephrotic syndrome in Japan using a national registry of clinical personal records. Sci Rep 2023; 13:14771. [PMID: 37679492 PMCID: PMC10485053 DOI: 10.1038/s41598-023-41909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023] Open
Abstract
The nationwide clinical features of Japanese patients with primary nephrotic syndrome (NS), including minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS), or membranous nephropathy (MN), have not yet been reported. We collected the clinical personal records of patients with primary NS between 2015 and 2018 from the national registry organized by the Japanese Ministry of Health, Labour, and Welfare. Overall, the demographics, chronic kidney disease classification based on glomerular filtration rate and albuminuria, and treatment of 6036 patients were collected: 3394 (56.2%) with MCD, 677 (11.2%) with FSGS, 1455 (24.1%) with MN, and 510 (8.5%) with others. MN patients were older than MCD and FSGS patients (67 vs. 42 and 47 years, respectively). Steroid-dependent NS or frequently relapsing NS was found in 70.2%, 40.5%, and 24.6%, whereas steroid-resistant NS was found in 6.4%, 36.0%, and 37.9% of patients in the MCD, FSGS, and MN, respectively. The present oral prednisolone use (mean dose, mg/day) was 87.2% (21.2), 80.9% (20.0), and 77.5% (18.8) of patients in the MCD, FSGS, and MN, respectively. The national registry of clinical personal records of primary NS could provide an informative insight into the characteristics, clinical features, and treatment approaches for patients with primary NS in Japan.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Division of Cardiology, Nephrology, Pulmonology and Neurology, Department of Internal Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-higashi, Asahikawa, Japan.
| | - Tomonori Kimura
- Reverse Translational Research Project, Center for Rare Disease Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
- Laboratory of Rare Disease Resource Library, Center for Rare Disease Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Ryuichi Sakate
- Reverse Translational Research Project, Center for Rare Disease Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Takehiko Wada
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
- Department of Nephrology, Toranomon Hospital, Tokyo, Japan
| | - Kengo Furuichi
- Department of Nephrology, Kanazawa Medical University School of Medicine, Uchinada, Japan
| | - Hirokazu Okada
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
109
|
Wearne N, Davidson B, Blockman M, Jones J, Ross IL, Dave JA. Management of Type 2 Diabetes Mellitus and Kidney Failure in People with HIV-Infection in Africa: Current Status and a Call to Action. HIV AIDS (Auckl) 2023; 15:519-535. [PMID: 37700755 PMCID: PMC10493098 DOI: 10.2147/hiv.s396949] [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: 06/23/2023] [Accepted: 08/13/2023] [Indexed: 09/14/2023] Open
Abstract
There is an increasing global burden of diabetes mellitus (DM) and chronic kidney disease (CKD), coupled with a high burden of people with HIV (PWH). Due to an increased lifespan on ART, PWH are now at risk of developing non-communicable diseases, including DM. Africa has the greatest burden of HIV infection and will experience the greatest increase in prevalence of DM over the next two decades. In addition, there is a rising number of people with CKD and progression to kidney failure. Therefore, there is an urgent need for the early identification and management of all 3 diseases to prevent disease progression and complications. This is particularly important in Africa for people with CKD where there is restricted or no access to dialysis and/or transplantation. This review focuses on the epidemiology and pathophysiology of the interaction between HIV infection and DM and the impact that these diseases have on the development and progression of CKD. Finally, it also aims to review the data on the management, which stems from the growing burden of all three diseases.
Collapse
Affiliation(s)
- Nicola Wearne
- Division of Nephrology and Hypertension, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - Bianca Davidson
- Division of Nephrology and Hypertension, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - Marc Blockman
- Division of Clinical Pharmacology, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - Jackie Jones
- Medicines Information Centre, Division of Clinical Pharmacology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ian L Ross
- Division of Endocrinology, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - Joel A Dave
- Division of Endocrinology, Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| |
Collapse
|
110
|
Pell J, Nagata S, Menon MC. Nonpodocyte Roles of APOL1 Variants: An Evolving Paradigm. KIDNEY360 2023; 4:e1325-e1331. [PMID: 37461136 PMCID: PMC10550003 DOI: 10.34067/kid.0000000000000216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
Since the seminal discovery of the trypanolytic, exonic variants in apolipoprotein L1 (APOL1) and their association with kidney disease in individuals of recent African ancestry, a wide body of research has emerged offering key insights into the mechanisms of disease. Importantly, the podocyte has become a focal point for our understanding of how risk genotype leads to disease, with activation of putative signaling pathways within the podocyte identified as playing a causal role in podocytopathy, FSGS, and progressive renal failure. However, the complete mechanism of genotype-to-phenotype progression remains incompletely understood in APOL1-risk individuals. An emerging body of evidence reports more than podocyte-intrinsic expression of APOL1 risk variants is needed for disease to manifest. This article reviews the seminal data and reports which placed the podocyte at the center of our understanding of APOL1-FSGS, as well as the evident shortcomings of this podocentric paradigm. We examine existing evidence for environmental and genetic factors that may influence disease, drawing from both clinical data and APOL1's fundamental role as an immune response gene. We also review the current body of data for APOL1's impact on nonpodocyte cells, including endothelial cells, the placenta, and immune cells in both a transplant and native setting. Finally, we discuss the implications of these emerging data and how the paradigm of disease might evolve as a result.
Collapse
Affiliation(s)
- John Pell
- Department of Medicine , Yale University, New Haven , Connecticut
| | | | | |
Collapse
|
111
|
Lea AJ, Clark AG, Dahl AW, Devinsky O, Garcia AR, Golden CD, Kamau J, Kraft TS, Lim YAL, Martins DJ, Mogoi D, Pajukanta P, Perry GH, Pontzer H, Trumble BC, Urlacher SS, Venkataraman VV, Wallace IJ, Gurven M, Lieberman DE, Ayroles JF. Applying an evolutionary mismatch framework to understand disease susceptibility. PLoS Biol 2023; 21:e3002311. [PMID: 37695771 PMCID: PMC10513379 DOI: 10.1371/journal.pbio.3002311] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/21/2023] [Indexed: 09/13/2023] Open
Abstract
Noncommunicable diseases (NCDs) are on the rise worldwide. Obesity, cardiovascular disease, and type 2 diabetes are among a long list of "lifestyle" diseases that were rare throughout human history but are now common. The evolutionary mismatch hypothesis posits that humans evolved in environments that radically differ from those we currently experience; consequently, traits that were once advantageous may now be "mismatched" and disease causing. At the genetic level, this hypothesis predicts that loci with a history of selection will exhibit "genotype by environment" (GxE) interactions, with different health effects in "ancestral" versus "modern" environments. To identify such loci, we advocate for combining genomic tools in partnership with subsistence-level groups experiencing rapid lifestyle change. In these populations, comparisons of individuals falling on opposite extremes of the "matched" to "mismatched" spectrum are uniquely possible. More broadly, the work we propose will inform our understanding of environmental and genetic risk factors for NCDs across diverse ancestries and cultures.
Collapse
Affiliation(s)
- Amanda J. Lea
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Andrew G. Clark
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, New York, United States of America
| | - Andrew W. Dahl
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Orrin Devinsky
- Department of Neurology, NYU Langone Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, New York, United States of America
| | - Angela R. Garcia
- Department of Anthropology, Stanford University, Stanford, California, United States of America
| | - Christopher D. Golden
- Department of Nutrition, Harvard T H Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Joseph Kamau
- One Health Centre, Institute of Primate Research, Karen, Nairobi, Kenya
| | - Thomas S. Kraft
- Department of Anthropology, University of Utah, Salt Lake City, Utah, United States of America
| | - Yvonne A. L. Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Dino J. Martins
- Turkana Basin Institute, Stony Brook University, Stony Brook, New York, United States of America
| | - Donald Mogoi
- Department of Medical Services and Public Health, Ministry of Health Laikipia County, Nanyuki, Kenya
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California, United States of America
| | - George H. Perry
- Departments of Anthropology and Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Herman Pontzer
- Department of Evolutionary Anthropology, Duke University, Durham, North Carolina, United States of America
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - Benjamin C. Trumble
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, United States of America
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, United States of America
| | - Samuel S. Urlacher
- Department of Anthropology, Baylor University, Waco, Texas, United States of America
| | - Vivek V. Venkataraman
- Department of Anthropology and Archaeology, University of Calgary, Calgary, Alberta, Canada
| | - Ian J. Wallace
- Department of Anthropology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Michael Gurven
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California, United States of America
| | - Daniel E. Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Julien F. Ayroles
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
| |
Collapse
|
112
|
Chen KW, Chang EL, Sheridan AM, Papaliodis GN. Tubulointerstitial nephritis and uveitis syndrome (TINU) following COVID-19 vaccination. Am J Ophthalmol Case Rep 2023; 31:101869. [PMID: 37332677 PMCID: PMC10257335 DOI: 10.1016/j.ajoc.2023.101869] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 05/01/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Affiliation(s)
- Kevin W Chen
- Drexel University College of Medicine, Philadelphia, PA, United States
| | - Eileen L Chang
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
- Stony Brook Medicine, Stony Brook, NY, United States
| | - Alice M Sheridan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - George N Papaliodis
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
113
|
Williams P. Retaining Race in Chronic Kidney Disease Diagnosis and Treatment. Cureus 2023; 15:e45054. [PMID: 37701164 PMCID: PMC10495104 DOI: 10.7759/cureus.45054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2023] [Indexed: 09/14/2023] Open
Abstract
The best overall measure of kidney function is glomerular filtration rate (GFR) as commonly estimated from serum creatinine concentrations (eGFRcr) using formulas that correct for the higher average creatinine concentrations in Blacks. After two decades of use, these formulas have come under scrutiny for estimating GFR differently in Blacks and non-Blacks. Discussions of whether to include race (Black vs. non-Black) in the calculation of eGFRcr fail to acknowledge that the original race-based eGFRcr provided the same CKD treatment recommendations for Blacks and non-Blacks based on directly (exogenously) measured GFR. Nevertheless, the National Kidney Foundation and the American Society of Nephrology Task Force on Reassessing the Inclusion of Race in Diagnosing Kidney Disease removed race in CKD treatment guidelines and pushed for the immediate adoption of a race-free eGFRcr formula by physicians and clinical laboratories. This formula is projected to negate CKD in 5.51 million White and other non-Black adults and reclassify CKD to less severe stages in another 4.59 million non-Blacks, in order to expand treatment eligibility to 434,000 Blacks not previously diagnosed and to 584,000 Blacks previously diagnosed with less severe CKD. This review examines: 1) the validity of the arguments for removing the original race correction, and 2) the performance of the proposed replacement formula. Excluding race in the derivation of eGFRcr changed the statistical bias from +3.7 to -3.6 ml/min/1.73m2 in Blacks and from +0.5 to +3.9 in non-Blacks, i.e., promoting CKD diagnosis in Blacks at the cost of restricting diagnosis in non-Blacks. By doing so, the revised eGFRcr greatly exaggerates the purported racial disparity in CKD burden. Claims that the revised formulas identify heretofore undiagnosed CKD in Blacks are not supported when studies that used kidney failure replacement therapy and mortality are interpreted as proxies for baseline CKD. Alternatively, a race-stratified eGFRcr (i.e., separate equations for Blacks and non-Blacks) would provide the least biased eGFRcr for both Blacks and non-Blacks and the best medical treatment for all patients.
Collapse
Affiliation(s)
- Paul Williams
- Life Sciences, Lawrence Berkeley National Laboratory, Berkeley, USA
| |
Collapse
|
114
|
Kalyesubula R, Wearne N, Kubo M, Hussey N, Naicker S. HIV and Associated TB: A Lethal Association for Kidney Health? Semin Nephrol 2023; 43:151470. [PMID: 38245391 DOI: 10.1016/j.semnephrol.2023.151470] [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: 01/22/2024]
Abstract
Human immunodeficiency virus (HIV) and tuberculosis (TB) are the leading infectious causes of death globally. The combined brunt of these diseases is experienced mainly in low-income and lower-middle-income countries. HIV/TB have devastating effects on the kidneys, leading to accelerated decline of kidney function as well as mortality. Managing the triad of TB/HIV and kidney disease is challenging. We discuss the epidemiology of HIV/TB coinfection and the kidney and the key mechanisms of kidney disease including genetic susceptibility. The clinical presentation and pathology, as well as the challenges of diagnosing CKD in these patients, also are discussed. The strategies to prevent and manage HIV/TB-related kidney disease such as proper assessment, avoiding nephrotoxic regimens, drug dose adjustments, kidney function monitoring, avoidance of drug-drug interactions, and other interventions are explored. We also briefly discuss the complexities around HIV/TB patients on dialysis and kidney transplantation. HIV/TB coinfection presents an increased risk for kidney-related morbidity and mortality; patients with this triad need to be given special consideration for future research and management.
Collapse
Affiliation(s)
- Robert Kalyesubula
- Department of Physiology and Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda.
| | - Nicola Wearne
- Division of Nephrology and Hypertension, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Mary Kubo
- Department of Clinical Medicine and Therapeutics, East African Kidney Institute, Faculty of Health Sciences, University of Nairobi, Nairobi, Kenya.
| | - Nadia Hussey
- Division of Nephrology and Hypertension, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Saraladevi Naicker
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
115
|
Chevalier RL. Evolution-informed therapy for kidney disease. Evol Med Public Health 2023; 11:316-317. [PMID: 37711226 PMCID: PMC10499305 DOI: 10.1093/emph/eoad027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/11/2023] [Indexed: 09/16/2023] Open
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
116
|
Birlutiu V, Neamtu B, Birlutiu RM, Ghibu AM, Dobritoiu ES. Our Experience with SARS-CoV-2 Infection and Acute Kidney Injury: Results from a Single-Center Retrospective Observational Study. Healthcare (Basel) 2023; 11:2402. [PMID: 37685436 PMCID: PMC10487568 DOI: 10.3390/healthcare11172402] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Renal failure in COVID-19 patients is reportedly related to multiple factors such as a direct SARS-CoV-2 cytopathic effect, cytokine storm, the association of pulmonary and/or cardiovascular lesions, the presence of thrombotic microangiopathy, endothelial damage, or the use of potentially nephrotoxic medications. METHODS We retrospectively analyzed 466 cases of SARS-CoV-2 infection, comparing 233 patients with acute kidney injury (AKI) with 233 patients without AKI in terms of their demographic characteristics, comorbidities, clinical background, laboratory investigations, time of AKI onset, therapy, and outcomes after using univariate analysis and a CART decision-tree approach. The latter was constructed in a reverse manner, starting from the top with the root and branching out until the splitting ceased, interconnecting all the predictors to predict the overall outcome (AKI vs. non-AKI). RESULTS There was a statistically significant difference between the clinical form distribution in the two groups, with fewer mild (2 vs. 5) and moderate (54 vs. 133) cases in the AKI group than in the non-AKI group and more severe and critical patients in the AKI cohort (116 vs. 92 and 60 vs. 3). There were four deaths (1.71%) in the non-AKI group and 120 deaths in the AKI group (51.5%) (p-value < 0.001). We noted statistically significant differences between the two study groups in relation to different tissue lesions (LDH), particularly at the pulmonary (CT severity score), hepatic (AST, ALT), and muscular levels (Creatine kinase). In addition, an exacerbated procoagulant and inflammatory profile in the study group was observed. The CART algorithm approach yielded decision paths that helped sort the risk of AKI progression into three categories: the low-risk category (0-40%), the medium-risk category (40-80%), and the high-risk category (>80%). It recognized specific inflammatory and renal biomarker profiles with particular cut-off points for procalcitonin, ferritin, LDH, creatinine, initial urea, and creatinine levels as important predictive factors of AKI outcomes (93.3% overall performance). CONCLUSIONS Our study revealed the association between particular risk factors and AKI progression in COVID-19 patients. Diabetes, dyspnea on admission, the need for supplemental oxygen, and admission to the intensive care unit all had a crucial role in producing unfavorable outcomes, with a death rate of more than 50%. Necessary imaging studies (CT scan severity score) and changes in specific biomarker levels (ferritin and C-reactive protein levels) were also noted. These factors should be further investigated in conjunction with the pathophysiological mechanisms of AKI progression in COVID-19 patients.
Collapse
Affiliation(s)
- Victoria Birlutiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- County Clinical Emergency Hospital, Bvd Corneliu Coposu, Nr. 2-4, 550245 Sibiu, Romania
| | - Bogdan Neamtu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- Pediatric Research Department, Pediatric Clinical Hospital Sibiu, Str. Pompeiu Onofreiu, Nr. 2-4, 550166 Sibiu, Romania
| | - Rares-Mircea Birlutiu
- Clinical Hospital of Orthopedics, Traumatology, and Osteoarticular TB Bucharest, B-dul Ferdinand 35–37, Sector 2, 021382 Bucharest, Romania
| | - Andreea Magdalena Ghibu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- County Clinical Emergency Hospital, Bvd Corneliu Coposu, Nr. 2-4, 550245 Sibiu, Romania
| | - Elena Simona Dobritoiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Romania, Str. Lucian Blaga, Nr. 2A, 550169 Sibiu, Romania
- County Clinical Emergency Hospital, Bvd Corneliu Coposu, Nr. 2-4, 550245 Sibiu, Romania
| |
Collapse
|
117
|
Gupta Y, Friedman DJ, McNulty M, Khan A, Lane B, Wang C, Ke J, Jin G, Wooden B, Knob AL, Lim TY, Appel GB, Huggins K, Liu L, Mitrotti A, Stangl MC, Bomback A, Westland R, Bodria M, Marasa M, Shang N, Cohen DJ, Crew RJ, Morello W, Canetta P, Radhakrishnan J, Martino J, Liu Q, Chung WK, Espinoza A, Luo Y, Wei WQ, Feng Q, Weng C, Fang Y, Kullo IJ, Naderian M, Limdi N, Irvin MR, Tiwari H, Mohan S, Rao M, Dube G, Chaudhary NS, Gutiérrez OM, Judd SE, Cushman M, Lange LA, Lange EM, Bivona DL, Verbitsky M, Winkler CA, Kopp JB, Santoriello D, Batal I, Brant Pinheiro SV, Araújo Oliveira E, E Silva ACS, Pisani I, Fiaccadori E, Lin F, Gesualdo L, Amoroso A, Ghiggeri GM, D'Agati VD, Magistroni R, Kenny EE, Loos RJF, Montini G, Hildebrandt F, Paul DS, Petrovski S, Goldstein DB, Kretzler M, Gbadegesin R, Gharavi AG, Kiryluk K, Sampson MG, Pollak MR, Sanna-Cherchi S. Strong protective effect of the APOL1 p.N264K variant against G2-associated focal segmental glomerulosclerosis and kidney disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.02.23293554. [PMID: 37577628 PMCID: PMC10418582 DOI: 10.1101/2023.08.02.23293554] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Black Americans have a significantly higher risk of developing chronic kidney disease (CKD), especially focal segmental glomerulosclerosis (FSGS), than European Americans. Two coding variants (G1 and G2) in the APOL1 gene play a major role in this disparity. While 13% of Black Americans carry the high-risk recessive genotypes, only a fraction of these individuals develops FSGS or kidney failure, indicating the involvement of additional disease modifiers. Here, we show that the presence of the APOL1 p.N264K missense variant, when co-inherited with the G2 APOL1 risk allele, substantially reduces the penetrance of the G1G2 and G2G2 high-risk genotypes by rendering these genotypes low-risk. These results align with prior functional evidence showing that the p.N264K variant reduces the toxicity of the APOL1 high-risk alleles. These findings have important implications for our understanding of the mechanisms of APOL1 -associated nephropathy, as well as for the clinical management of individuals with high-risk genotypes that include the G2 allele.
Collapse
|
118
|
Cai Y, Liu Y, Tong J, Jin Y, Liu J, Hao X, Ji Y, Ma J, Pan X, Chen N, Ren H, Xie J. Develop and Validate a Risk Score in Predicting Renal Failure in Focal Segmental Glomerulosclerosis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:285-297. [PMID: 37899999 PMCID: PMC10601954 DOI: 10.1159/000529773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/08/2023] [Indexed: 10/31/2023]
Abstract
Introduction The aim of this study was to develop and validate a risk score (RS) for end-stage kidney disease (ESKD) in patients with focal segmental glomerulosclerosis (FSGS). Methods Patient with biopsy-proven FSGS was enrolled. All the patients were allocated 1:1 to the two groups according to their baseline gender, age, and baseline creatinine level by using a stratified randomization method. ESKD was the primary endpoint. Results We recruited 359 FSGS patients, and 177 subjects were assigned to group 1 and 182 to group 2. The clinicopathological variables were similar between two groups. There were 23 (13%) subjects reached to ESKD in group 1 and 22 (12.1%) in group 2. By multivariate Cox regression analyses, we established RS 1 and RS 2 in groups 1 and 2, respectively. RS 1 consists of five parameters including lower eGFR, higher urine protein, MAP, IgG level, and tubulointerstitial lesion (TIL) score; RS 2 also consists of five predictors including lower C3, higher MAP, IgG level, hemoglobin, and TIL score. RS 1 and RS 2 were cross-validated between these two groups, showing RS 1 had better performance in predicting 5-year ESKD in group 1 (c statics, 0.86 [0.74-0.98] vs. 0.82 [0.69-0.95]) and group 2 (c statics, 0.91 [0.83-0.99] vs. 0.89 [0.79-0.99]) compared to RS 2. We then stratified the risk factors into four groups, and Kaplan-Meier survival curve revealed that patients progressed to ESKD increased as risk levels increased. Conclusions A predictive model incorporated clinicopathological feature was developed and validated for the prediction of ESKD in FSGS patients.
Collapse
Affiliation(s)
- Yikai Cai
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunzi Liu
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Tong
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanmeng Jin
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Liu
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Hao
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinhong Ji
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Ma
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxia Pan
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Chen
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Ren
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyuan Xie
- Department of Nephrology, Institute of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
119
|
Marasa M, Ahram DF, Rehman AU, Mitrotti A, Abhyankar A, Jain NG, Weng PL, Piva SE, Fernandez HE, Uy NS, Chatterjee D, Kil BH, Nestor JG, Felice V, Robinson D, Whyte D, Gharavi AG, Appel GB, Radhakrishnan J, Santoriello D, Bomback A, Lin F, D’Agati VD, Jobanputra V, Sanna-Cherchi S. Implementation and Feasibility of Clinical Genome Sequencing Embedded Into the Outpatient Nephrology Care for Patients With Proteinuric Kidney Disease. Kidney Int Rep 2023; 8:1638-1647. [PMID: 37547535 PMCID: PMC10403677 DOI: 10.1016/j.ekir.2023.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/01/2023] [Accepted: 05/22/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction The diagnosis and management of proteinuric kidney diseases such as focal segmental glomerulosclerosis (FSGS) are challenging. Genetics holds the promise to improve clinical decision making for these diseases; however, it is often performed too late to enable timely clinical action and it is not implemented within routine outpatient nephrology visits. Methods We sought to test the implementation and feasibility of clinical rapid genome sequencing (GS) in guiding decision making in patients with proteinuric kidney disease in real-time and embedded in the outpatient nephrology setting. Results We enrolled 10 children or young adults with biopsy-proven FSGS (9 cases) or minimal change disease (1 case). The mean age at enrollment was 16.2 years (range 2-30). The workflow did not require referral to external genetics clinics but was conducted entirely during the nephrology standard-of-care appointments. The total turn-around-time from enrollment to return-of-results and clinical decision averaged 21.8 days (12.4 for GS), which is well within a time frame that allows clinically relevant treatment decisions. A monogenic or APOL1-related form of kidney disease was diagnosed in 5 of 10 patients. The genetic findings resulted in a rectified diagnosis in 6 patients. Both positive and negative GS findings determined a change in pharmacological treatment. In 3 patients, the results were instrumental for transplant evaluation, donor selection, and the immunosuppressive treatment. All patients and families received genetic counseling. Conclusion Clinical GS is feasible and can be implemented in real-time in the outpatient care to help guiding clinical management. Additional studies are needed to confirm the cost-effectiveness and broader utility of clinical GS across the phenotypic and demographic spectrum of kidney diseases.
Collapse
Affiliation(s)
- Maddalena Marasa
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Dina F. Ahram
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | | | - Adele Mitrotti
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | | | - Namrata G. Jain
- Division of Pediatric Nephrology, Department of Pediatrics, Columbia University, New York, USA
| | - Patricia L. Weng
- Division of Pediatric Nephrology, Department of Pediatrics, UCLA Medical Center and UCLA Medical Center-Santa Monica, Los Angeles, California, USA
| | - Stacy E. Piva
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Hilda E. Fernandez
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Natalie S. Uy
- Division of Pediatric Nephrology, Department of Pediatrics, Columbia University, New York, USA
| | - Debanjana Chatterjee
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Byum H. Kil
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Jordan G. Nestor
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | | | | | - Dilys Whyte
- Pediatric Specialty Center of Good Samaritan Hospital Medical Center, Babylon, New York, USA
| | - Ali G. Gharavi
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Gerald B. Appel
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Jai Radhakrishnan
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Dominick Santoriello
- Department of Pathology and Cell Biology, Renal Pathology Division, Columbia University Medical Center, New York, USA
| | - Andrew Bomback
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| | - Fangming Lin
- Division of Pediatric Nephrology, Department of Pediatrics, Columbia University, New York, USA
| | - Vivette D. D’Agati
- Department of Pathology and Cell Biology, Renal Pathology Division, Columbia University Medical Center, New York, USA
| | - Vaidehi Jobanputra
- The New York Genome Center, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA
| |
Collapse
|
120
|
Parr MF, Hidalgo G, Goldstein MJ, Batal I, Lieberman KV, Amoruso MR, Baer AZ, Jain NG. CMV-associated collapsing focal segmental glomerulosclerosis after kidney transplant in a pediatric patient. Pediatr Transplant 2023; 27:e14535. [PMID: 37128132 PMCID: PMC10753975 DOI: 10.1111/petr.14535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/24/2023] [Accepted: 04/13/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Cytomegalovirus (CMV) is a significant cause of morbidity among immunocompromised patients who have undergone kidney transplantation and is known to rarely induce collapsing focal segmental glomerulosclerosis (FSGS) among adults. METHODS We present the first reported case of CMV-induced collapsing FSGS in a pediatric patient after kidney transplant. RESULTS Our patient underwent a deceased donor kidney transplant due to end-stage renal disease secondary to lupus nephritis. Approximately 4 months after transplantation, he developed signs of worsening kidney function in the setting of CMV viremia and was found to have collapsing features of FSGS on kidney transplant biopsy. He was managed with a prompt escalation of antiviral therapy along with a reduction of immunosuppression and recovered without significant complication. At follow-up, he continued to have undetectable CMV titers, creatinine within normal limits, and no significant proteinuria. CONCLUSION This report demonstrates CMV as a cause of collapsing FSGS and should be considered among pediatric transplant recipients who present with acute kidney injury, as should early assessment of APOL1 genetic status in both donor and recipient.
Collapse
Affiliation(s)
- Madeline F.E. Parr
- Department of Pediatrics, Division of Pediatric Nephrology, Joseph M. Sanzari Children’s Hospital at Hackensack University Medical Center, Hackensack, NJ
| | - Guillermo Hidalgo
- Department of Pediatrics, Division of Pediatric Nephrology, Jersey Shore University Medical Center, Neptune, NJ
| | - Michael J. Goldstein
- Department of Transplant Surgery, Hackensack University Medical Center, Hackensack, NJ
| | - Ibrahim Batal
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Kenneth V. Lieberman
- Department of Pediatrics, Division of Pediatric Nephrology, Joseph M. Sanzari Children’s Hospital at Hackensack University Medical Center, Hackensack, NJ
| | - Marlene R. Amoruso
- Department of Transplant Surgery, Hackensack University Medical Center, Hackensack, NJ
| | - Aryeh Z. Baer
- Department of Pediatrics, Division of Pediatric Infectious Disease, Joseph M Sanzari Children’s Hospital at Hackensack University Medical Center, Hackensack, NJ
| | - Namrata G. Jain
- Department of Pediatrics, Division of Pediatric Nephrology, Joseph M. Sanzari Children’s Hospital at Hackensack University Medical Center, Hackensack, NJ
| |
Collapse
|
121
|
Nkoy AB, Ekulu PM, Labarque V, Van den Heuvel LP, Levtchenko EN. HIV-associated nephropathy in children: challenges in a resource-limited setting. Pediatr Nephrol 2023; 38:2509-2521. [PMID: 36472655 DOI: 10.1007/s00467-022-05819-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
HIV infection remains one of the leading causes of morbidity and mortality worldwide, especially in children living in resource-limited settings. Although the World Health Organization (WHO) recently recommended antiretroviral therapy (ART) initiation upon diagnosis regardless of the number of CD4, ART access remains limited, especially in children living in sub-Saharan Africa (SSA). HIV-infected children who do not receive appropriate ART are at increased risk of developing HIV-associated nephropathy (HIVAN). Although due to genetic susceptibility, SSA is recognized to be the epicenter of HIVAN, limited information is available regarding the burden of HIVAN in children living in Africa. The present review discusses the information available to date on the prevalence, pathogenesis, risk factors, diagnosis, and management of HIVAN in children, focusing on related challenges in a resource-limited setting.
Collapse
Affiliation(s)
- Agathe B Nkoy
- Division of Nephrology, Department of Pediatrics, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of Congo
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Pépé M Ekulu
- Division of Nephrology, Department of Pediatrics, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Veerle Labarque
- Department of Pediatric Hematology, University Hospital Leuven, Leuven, Belgium
- Center of Molecular and Vascular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lambertus P Van den Heuvel
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Pediatric Nephrology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Elena N Levtchenko
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, Katholieke Universiteit Leuven, Leuven, Belgium.
- Department of Pediatric Nephrology, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
122
|
Adebayo OC, Nkoy AB, van den Heuvel LP, Labarque V, Levtchenko E, Delanaye P, Pottel H. Glomerular hyperfiltration: part 2-clinical significance in children. Pediatr Nephrol 2023; 38:2529-2547. [PMID: 36472656 DOI: 10.1007/s00467-022-05826-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022]
Abstract
Glomerular hyperfiltration (GHF) is a phenomenon that can occur in various clinical conditions affecting the kidneys such as sickle cell disease, diabetes mellitus, autosomal dominant polycystic kidney disease, and solitary functioning kidney. Yet, the pathophysiological mechanisms vary from one disease to another and are not well understood. More so, it has been demonstrated that GHF may occur at the single-nephron in some clinical conditions while in others at the whole-kidney level. In this review, we explore the pathophysiological mechanisms of GHF in relation to various clinical conditions in the pediatric population. In addition, we discuss the role and mechanism of action of important factors such as gender, low birth weight, and race in the pathogenesis of GHF. Finally, in this current review, we further highlight the consequences of GHF in the progression of kidney disease.
Collapse
Affiliation(s)
- Oyindamola C Adebayo
- Center of Vascular and Molecular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Campus Gasthiusberg, 3000 Leuven, Belgium
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, Katholieke Universiteit Leuven, Campus Gasthiusberg, 3000 Leuven, Belgium
| | - Agathe B Nkoy
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, Katholieke Universiteit Leuven, Campus Gasthiusberg, 3000 Leuven, Belgium
- Division of Nephrology, Department of Pediatrics, Faculty of Medicine, University Hospital of Kinshasa, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Lambertus P van den Heuvel
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, Katholieke Universiteit Leuven, Campus Gasthiusberg, 3000 Leuven, Belgium
- Department of Pediatric Nephrology, Radboud University Medical Centre, 6500 Nijmegen, The Netherlands
| | - Veerle Labarque
- Center of Vascular and Molecular Biology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Campus Gasthiusberg, 3000 Leuven, Belgium
- Department of Pediatric Hematology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Elena Levtchenko
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, Katholieke Universiteit Leuven, Campus Gasthiusberg, 3000 Leuven, Belgium
- Department of Pediatric Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University of Liège, CHU Sart Tilman, Liège, Belgium
- Department of Nephrology-Dialysis-Apheresis, Hôpital Universitaire Carémeau, Nîmes, France
| | - Hans Pottel
- Department of Public Health and Primary Care, Katholieke Universiteit Leuven, Campus Kulak, 8500 Kortrijk, Belgium.
| |
Collapse
|
123
|
Gbadegesin R, Lane B. Inaxaplin for the treatment of APOL1-associated kidney disease. Nat Rev Nephrol 2023; 19:479-480. [PMID: 37106136 PMCID: PMC10461697 DOI: 10.1038/s41581-023-00721-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Chronic kidney disease (CKD) is highly prevalent World-Wide and it is an important cause of morbidity and mortality. In 2010, variants in apolipoprotein type 1 (APOL1) gene was identified as a major risk factor for higher prevalence of CKD in individuals of African ancestry. The mechanisms by which toxic gain of function APOL1 variants cause disease are the subjects of current investigations, and there is currently no effective targeted therapy for APOL1 associated kidney disease. Egbuna and others recently reported that an orally administered small molecule compound inaxaplin (VX-147) that binds APOL1 may be useful in the treatment of APOL1 associated kidney disease. This is a groundbreaking study, if confirmed in randomized control studies it may turn out to be one of the major advances in the therapy of proteinuric CKD this decade, and may also represent precision therapy for addressing health disparity in CKD. However, there is a need for a larger randomized control studies with stable eGFR and complete remission of proteinuria as end point, such studies should also be carried out in a more geographically diverse population.
Collapse
Affiliation(s)
- Rasheed Gbadegesin
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, USA.
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
| | - Brandon Lane
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
124
|
Stockwell AD, Chang MC, Mahajan A, Forrest W, Anegondi N, Pendergrass RK, Selvaraj S, Reeder J, Wei E, Iglesias VA, Creps NM, Macri L, Neeranjan AN, van der Brug MP, Scales SJ, McCarthy MI, Yaspan BL. Multi-ancestry GWAS analysis identifies two novel loci associated with diabetic eye disease and highlights APOL1 as a high risk locus in patients with diabetic macular edema. PLoS Genet 2023; 19:e1010609. [PMID: 37585454 PMCID: PMC10461827 DOI: 10.1371/journal.pgen.1010609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 08/28/2023] [Accepted: 06/11/2023] [Indexed: 08/18/2023] Open
Abstract
Diabetic retinopathy (DR) is a common complication of diabetes. Approximately 20% of DR patients have diabetic macular edema (DME) characterized by fluid leakage into the retina. There is a genetic component to DR and DME risk, but few replicable loci. Because not all DR cases have DME, we focused on DME to increase power, and conducted a multi-ancestry GWAS to assess DME risk in a total of 1,502 DME patients and 5,603 non-DME controls in discovery and replication datasets. Two loci reached GWAS significance (p<5x10-8). The strongest association was rs2239785, (K150E) in APOL1. The second finding was rs10402468, which co-localized to PLVAP and ANKLE1 in vascular / endothelium tissues. We conducted multiple sensitivity analyses to establish that the associations were specific to DME status and did not reflect diabetes status or other diabetic complications. Here we report two novel loci for risk of DME which replicated in multiple clinical trial and biobank derived datasets. One of these loci, containing the gene APOL1, is a risk factor in African American DME and DKD patients, indicating that this locus plays a broader role in diabetic complications for multiple ancestries. Trial Registration: NCT00473330, NCT00473382, NCT03622580, NCT03622593, NCT04108156.
Collapse
Affiliation(s)
| | | | - Anubha Mahajan
- Genentech, San Francisco, California, United States of America
| | - William Forrest
- Genentech, San Francisco, California, United States of America
| | - Neha Anegondi
- Genentech, San Francisco, California, United States of America
| | | | - Suresh Selvaraj
- Genentech, San Francisco, California, United States of America
| | - Jens Reeder
- Genentech, San Francisco, California, United States of America
| | - Eric Wei
- Genentech, San Francisco, California, United States of America
| | | | | | - Laura Macri
- Character Biosciences, San Francisco, California, United States of America
| | | | | | - Suzie J. Scales
- Genentech, San Francisco, California, United States of America
| | | | - Brian L. Yaspan
- Genentech, San Francisco, California, United States of America
| |
Collapse
|
125
|
Olabisi OA. APOL1 channel blocker reduces proteinuria in FSGS. Kidney Int 2023; 104:228-230. [PMID: 37224918 DOI: 10.1016/j.kint.2023.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/26/2023]
Affiliation(s)
- Opeyemi A Olabisi
- Department of Medicine, Division of Nephrology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
126
|
Zaidi AA, Verma A, Morse C, Ritchie MD, Mathieson I. The genetic and phenotypic correlates of mtDNA copy number in a multi-ancestry cohort. HGG ADVANCES 2023; 4:100202. [PMID: 37255673 PMCID: PMC10225932 DOI: 10.1016/j.xhgg.2023.100202] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/25/2023] [Indexed: 06/01/2023] Open
Abstract
Mitochondrial DNA copy number (mtCN) is often treated as a proxy for mitochondrial (dys-) function and disease risk. Pathological changes in mtCN are common symptoms of rare mitochondrial disorders, but reported associations between mtCN and common diseases vary across studies. To understand the biology of mtCN, we carried out genome- and phenome-wide association studies of mtCN in 30,666 individuals from the Penn Medicine BioBank (PMBB)-a diverse cohort of largely African and European ancestry. We estimated mtCN in peripheral blood using exome sequence data, taking cell composition into account. We replicated known genetic associations of mtCN in the PMBB and found that their effects are highly correlated between individuals of European and African ancestry. However, the heritability of mtCN was much higher among individuals of largely African ancestry ( h 2 = 0.3 ) compared with European ancestry individuals( h 2 = 0.1 ) . Admixture mapping suggests that there are undiscovered variants underlying mtCN that are differentiated in frequency between individuals with African and European ancestry. We show that mtCN is associated with many health-related phenotypes. We discovered robust associations between mtDNA copy number and diseases of metabolically active tissues, such as cardiovascular disease and liver damage, that were consistent across African and European ancestry individuals. Other associations, such as epilepsy and prostate cancer, were only discovered in either individuals with European or African ancestry but not both. We show that mtCN-phenotype associations can be sensitive to blood cell composition and environmental modifiers, explaining why such associations are inconsistent across studies. Thus, mtCN-phenotype associations must be interpreted with care.
Collapse
Affiliation(s)
- Arslan A. Zaidi
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anurag Verma
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colleen Morse
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Penn Medicine BioBank
- Center for Translational Bioinformatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Iain Mathieson
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
127
|
Vincenti F, Angeletti A, Ghiggeri GM. State of the art in childhood nephrotic syndrome: concrete discoveries and unmet needs. Front Immunol 2023; 14:1167741. [PMID: 37503337 PMCID: PMC10368981 DOI: 10.3389/fimmu.2023.1167741] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Nephrotic syndrome (NS) is a clinical entity characterized by proteinuria, hypoalbuminemia, and peripheral edema. NS affects about 2-7 per 100,000 children aged below 18 years old yearly and is classified, based on the response to drugs, into steroid sensitive (SSNS), steroid dependent, (SDNS), multidrug dependent (MDNS), and multidrug resistant (MRNS). Forms of NS that are more difficult to treat are associated with a worse outcome with respect to renal function. In particular, MRNS commonly progresses to end stage renal failure requiring renal transplantation, with recurrence of the original disease in half of the cases. Histological presentations of NS may vary from minimal glomerular lesions (MCD) to focal segmental glomerulosclerosis (FSGS) and, of relevance, the histological patterns do not correlate with the response to treatments. Moreover, around half of MRNS cases are secondary to causative pathogenic variants in genes involved in maintaining the glomerular structure. The pathogenesis of NS is still poorly understood and therapeutic approaches are mostly based on clinical experience. Understanding of pathogenetic mechanisms of NS is one of the 'unmet needs' in nephrology and represents a significant challenge for the scientific community. The scope of the present review includes exploring relevant findings, identifying unmet needs, and reviewing therapeutic developments that characterize NS in the last decades. The main aim is to provide a basis for new perspectives and mechanistic studies in NS.
Collapse
Affiliation(s)
- Flavio Vincenti
- Division of Nephrology, Department of Medicine and Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Andrea Angeletti
- Nephrology Dialysis and Transplantation, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Gian Marco Ghiggeri
- Nephrology Dialysis and Transplantation, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
128
|
Riley R, Mathieson I, Mathieson S. INTERPRETING GENERATIVE ADVERSARIAL NETWORKS TO INFER NATURAL SELECTION FROM GENETIC DATA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531546. [PMID: 36945387 PMCID: PMC10028936 DOI: 10.1101/2023.03.07.531546] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Understanding natural selection in humans and other species is a major focus for the use of machine learning in population genetics. Existing methods rely on computationally intensive simulated training data. Unlike efficient neutral coalescent simulations for demographic inference, realistic simulations of selection typically requires slow forward simulations. Because there are many possible modes of selection, a high dimensional parameter space must be explored, with no guarantee that the simulated models are close to the real processes. Mismatches between simulated training data and real test data can lead to incorrect inference. Finally, it is difficult to interpret trained neural networks, leading to a lack of understanding about what features contribute to classification. Here we develop a new approach to detect selection that requires relatively few selection simulations during training. We use a Generative Adversarial Network (GAN) trained to simulate realistic neutral data. The resulting GAN consists of a generator (fitted demographic model) and a discriminator (convolutional neural network). For a genomic region, the discriminator predicts whether it is "real" or "fake" in the sense that it could have been simulated by the generator. As the "real" training data includes regions that experienced selection and the generator cannot produce such regions, regions with a high probability of being real are likely to have experienced selection. To further incentivize this behavior, we "fine-tune" the discriminator with a small number of selection simulations. We show that this approach has high power to detect selection in simulations, and that it finds regions under selection identified by state-of-the art population genetic methods in three human populations. Finally, we show how to interpret the trained networks by clustering hidden units of the discriminator based on their correlation patterns with known summary statistics. In summary, our approach is a novel, efficient, and powerful way to use machine learning to detect natural selection.
Collapse
Affiliation(s)
- Rebecca Riley
- Department of Computer Science, Haverford College, Haverford PA, 19041 USA
| | - Iain Mathieson
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia PA, 19104 USA
| | - Sara Mathieson
- Department of Computer Science, Haverford College, Haverford PA, 19041 USA
| |
Collapse
|
129
|
Williams AE, Esezobor CI, Lane BM, Gbadegesin RA. Hiding in plain sight: genetics of childhood steroid-resistant nephrotic syndrome in Sub-Saharan Africa. Pediatr Nephrol 2023; 38:2003-2012. [PMID: 36459247 PMCID: PMC10416081 DOI: 10.1007/s00467-022-05831-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is the most severe form of childhood nephrotic syndrome with an increased risk of progression to chronic kidney disease stage 5. Research endeavors to date have identified more than 80 genes that are associated with SRNS. Most of these genes regulate the structure and function of the podocyte, the visceral epithelial cells of the glomerulus. Although individuals of African ancestry have the highest prevalence of SRNS, especially those from Sub-Saharan Africa (SSA), with rates as high as 30-40% of all cases of nephrotic syndrome, studies focusing on the characterization and understanding of the genetic basis of SRNS in the region are negligible compared with Europe and North America. Therefore, it remains unclear if some of the variants in SRNS genes that are deemed pathogenic for SRNS are truly disease causing, and if the leading causes of monogenic nephrotic syndrome in other populations are the same for children in SSA with SRNS. Other implications of this lack of genetic data for SRNS in the region include the exclusion of children from the region from clinical trials aimed at identifying potential novel therapeutic agents for this severe form of nephrotic syndrome. This review underlines a need for concerted efforts to advance the genetic basis of SRNS in children in SSA. Such endeavors will complement global efforts at understanding the genetic basis of nephrotic syndrome.
Collapse
Affiliation(s)
- Anna Elizabeth Williams
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Christopher I Esezobor
- Department of Pediatrics, Faculty of Clinical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Brandon M Lane
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, 27710, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Rasheed A Gbadegesin
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, 27710, USA.
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.
| |
Collapse
|
130
|
Heras Benito M, Pérez García ML, Antúnez Plaza P, Montero Mateos E. [Unexpected diagnosis of nephronopthisis in the genetic study of hypertension due to histological diagnosis of benign nephroangioesclerosis evolved in a young caucasian patient]. HIPERTENSION Y RIESGO VASCULAR 2023; 40:150-153. [PMID: 36894476 DOI: 10.1016/j.hipert.2023.02.001] [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: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 03/09/2023]
Abstract
We present the case of a young Caucasian patient with renal disease of unclear cause, with a final diagnosis of advanced benign nephroangiosclerosis established by renal biopsy. Due to the possibility of having hypertension in pediatric age (without study or treatment), with the renal biopsy findings, the genetic study showed polymorphisms risk in the APOL1 and MYH9, and also an unexpected diagnosis of a complete deletion of the NPHP1 gene in homozygosis, associated with the development of nephronophthisis. In conclusion, this case illustrates the importance of carrying out a genetic study in youngs patients with renal disease unclear cause, even having a histological diagnosis of nephroangiosclerosis.
Collapse
Affiliation(s)
- M Heras Benito
- Servicio de Nefrología, Hospital Universitario de Salamanca, Salamanca, España.
| | - M L Pérez García
- Servicio de Medicina Interna, Hospital Universitario de Salamanca, Salamanca, España
| | - P Antúnez Plaza
- Servicio de Anatomía Patológica, Hospital Universitario de Salamanca, Salamanca, España
| | - E Montero Mateos
- Servicio de Anatomía Patológica, Hospital Universitario de Salamanca, Salamanca, España
| |
Collapse
|
131
|
Anders HJ, Kitching AR, Leung N, Romagnani P. Glomerulonephritis: immunopathogenesis and immunotherapy. Nat Rev Immunol 2023; 23:453-471. [PMID: 36635359 PMCID: PMC9838307 DOI: 10.1038/s41577-022-00816-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 01/14/2023]
Abstract
'Glomerulonephritis' (GN) is a term used to describe a group of heterogeneous immune-mediated disorders characterized by inflammation of the filtration units of the kidney (the glomeruli). These disorders are currently classified largely on the basis of histopathological lesion patterns, but these patterns do not align well with their diverse pathological mechanisms and hence do not inform optimal therapy. Instead, we propose grouping GN disorders into five categories according to their immunopathogenesis: infection-related GN, autoimmune GN, alloimmune GN, autoinflammatory GN and monoclonal gammopathy-related GN. This categorization can inform the appropriate treatment; for example, infection control for infection-related GN, suppression of adaptive immunity for autoimmune GN and alloimmune GN, inhibition of single cytokines or complement factors for autoinflammatory GN arising from inborn errors in innate immunity, and plasma cell clone-directed or B cell clone-directed therapy for monoclonal gammopathies. Here we present the immunopathogenesis of GN and immunotherapies in use and in development and discuss how an immunopathogenesis-based GN classification can focus research, and improve patient management and teaching.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University Munich, Munich, Germany.
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Department of Paediatric Nephrology, Monash Health, Clayton, VIC, Australia
| | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Paola Romagnani
- Department of Experimental and Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| |
Collapse
|
132
|
Hansrivijit P, Samal L, Mendu ML. Unpacking Contributors to CKD Incidence and Progression: Time to Move Beyond Traditional Risk Factors. Am J Kidney Dis 2023; 82:1-3. [PMID: 37061914 DOI: 10.1053/j.ajkd.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/17/2023] [Indexed: 04/17/2023]
Affiliation(s)
- Panupong Hansrivijit
- Nephrology Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lipika Samal
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts
| | - Mallika L Mendu
- Nephrology Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Office of the Chief Medical Officer, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
133
|
Ozeki T, Gillespie BW, Larkina M, Maruyama S, Alakwaa F, Kretzler M, Mariani LH. Clinical Course of Adult FSGS and Minimal Change Disease in North American and Japanese Cohorts. KIDNEY360 2023; 4:924-934. [PMID: 37131280 PMCID: PMC10371276 DOI: 10.34067/kid.0000000000000133] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/03/2023] [Indexed: 05/04/2023]
Abstract
Key Points Data from different geographical regions highlighted the differences in clinical manifestations and treatment response of adult FSGS/minimal change disease. There were shared factors that were associated with treatment response across the cohorts: FSGS, higher BP, and lower eGFR. Recognizing geographical difference allows us better understanding of disease biology, risk prediction, and design of future clinical trials. Background Regional differences in presentation and clinical course of nephrotic syndrome (NS) have not been studied well because few studies directly compared the data from different intercontinental regions. Methods We included adult nephrotic patients with FSGS and minimal change disease (MCD) who received immunosuppressive therapy (IST) in a North American (Nephrotic Syndrome Study Network [NEPTUNE], N =89) or Japanese (Nagoya Kidney Disease Registry [N-KDR], N =288) cohort. Baseline characteristics and rates of complete remission (CR) were compared. Factors associated with time to CR were evaluated by Cox regression models. Results NEPTUNE participants had more FSGS (53.9 versus 17.0%) and family history of kidney disease (35.2 versus 3.2%). N-KDR participants were older (median 56 versus 43 years) and demonstrated greater levels of urine protein creatinine ratio (7.73 versus 6.65) and hypoalbuminemia (1.6 versus 2.2 mg/dl). N-KDR participants showed higher proportion of CR (overall: 89.2 versus 62.9%; FSGS: 67.3 versus 43.7%; MCD: 93.7 versus 85.4%). A multivariable model showed that FSGS (versus MCD: hazard ratio [HR], 0.28; 95% confidence interval [CI], 0.20 to 0.41), systolic BP (per 10 mm Hg: HR, 0.93; 95% CI, 0.86 to 0.99), and eGFR (per 10 ml/min per 1.73 m2: HR, 1.16; 95% CI, 1.09 to 1.24) were associated with time to CR. There were significant interactions in patient age (P = 0.004) and eGFR (P = 0.001) between the cohorts. Conclusions The North American cohort had more FSGS and more frequent family history. Japanese patients showed more severe NS with better response to IST. FSGS, hypertension, and lower eGFR were shared predictors of poor treatment response. Identifying shared and unique features across geographically diverse populations may help uncover biologically relevant subgroups, improve prediction of disease course, and better design future multinational clinical trials.
Collapse
Affiliation(s)
- Takaya Ozeki
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Brenda W. Gillespie
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Maria Larkina
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fadhl Alakwaa
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Laura H. Mariani
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
134
|
Aklilu AM. Diagnosis of Chronic Kidney Disease and Assessing Glomerular Filtration Rate. Med Clin North Am 2023; 107:641-658. [PMID: 37258004 DOI: 10.1016/j.mcna.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Chronic kidney disease (CKD) is a silent progressive disease. It is diagnosed by assessing filtration and markers of kidney damage such as albuminuria. The diagnosis of CKD should include not only assessing the glomerular filtration rate (GFR) and albuminuria but also the cause. The CKD care plan should include documentation of the trajectory and prognosis. The use of a combination of serum cystatin C and creatinine concentration offers a more accurate estimation of GFR. Social determinants of health are important to address as part of the diagnosis because they contribute to CKD disparities.
Collapse
Affiliation(s)
- Abinet M Aklilu
- Section of Nephrology, Department of Medicine, Yale school of Medicine, 60 Temple Street, Suite 6C, New Haven, CT 06510, USA.
| |
Collapse
|
135
|
Rashmi P, Sigdel TK, Rychkov D, Damm I, Da Silva AA, Vincenti F, Lourenco AL, Craik CS, Reiser J, Sarwal MM. Perturbations in podocyte transcriptome and biological pathways induced by FSGS associated circulating factors. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:315. [PMID: 37404982 PMCID: PMC10316099 DOI: 10.21037/atm-22-3670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 12/03/2022] [Indexed: 07/06/2023]
Abstract
Background Focal segmental glomerulosclerosis (FSGS) is frequently associated with heavy proteinuria and progressive renal failure requiring dialysis or kidney transplantation. However, primary FSGS also has a ~40% risk of recurrence of disease in the transplanted kidney (rFSGS). Multiple circulating factors have been identified to contribute to the pathogenesis of primary and rFSGS including soluble urokinase-type plasminogen activator receptor (suPAR) and patient-derived CD40 autoantibody (CD40autoAb). However, the downstream effector pathways specific to individual factors require further study. The tumor necrosis factor, TNF pathway activation by one or more circulating factors present in the sera of patients with FSGS has been supported by multiple studies. Methods A human in vitro model was used to study podocyte injury measured as the loss of actin stress fibers. Anti-CD40 autoantibody was isolated from FSGS patients (recurrent and non-recurrent) and control patients with ESRD due to non-FSGS related causes. Two novel human antibodies-anti-uPAR (2G10) and anti-CD40 antibody (Bristol Meyer Squibb, 986090) were tested for their ability to rescue podocyte injury. Podocytes treated with patient derived antibody were transcriptionally profiled using whole human genome microarray. Results Here we show that podocyte injury caused by sera from FSGS patients is mediated by CD40 and suPAR and can be blocked by human anti-uPAR and anti-CD40 antibodies. Transcriptomic studies to compare the molecules and pathways activated in response to CD40 autoantibody from rFSGS patients (rFSGS/CD40autoAb) and suPAR, identified unique inflammatory pathways associated with FSGS injury. Conclusions We identified several novel and previously described genes associated with FSGS progression. Targeted blockade of suPAR and CD40 pathways with novel human antibodies showed inhibition of podocyte injury in FSGS.
Collapse
Affiliation(s)
- Priyanka Rashmi
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Tara K. Sigdel
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Dmitry Rychkov
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Izabella Damm
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Andrea Alice Da Silva
- Department of Immunology, Laboratory of Autoimmunity and Immunoregulation, Fluminense Federal University, Niteroi, Brazil
| | - Flavio Vincenti
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Andre L. Lourenco
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Charles S. Craik
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Minnie M. Sarwal
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
136
|
Verma A, Huffman JE, Rodriguez A, Conery M, Liu M, Ho YL, Kim Y, Heise DA, Guare L, Panickan VA, Garcon H, Linares F, Costa L, Goethert I, Tipton R, Honerlaw J, Davies L, Whitbourne S, Cohen J, Posner DC, Sangar R, Murray M, Wang X, Dochtermann DR, Devineni P, Shi Y, Nandi TN, Assimes TL, Brunette CA, Carroll RJ, Clifford R, Duvall S, Gelernter J, Hung A, Iyengar SK, Joseph J, Kember R, Kranzler H, Levey D, Luoh SW, Merritt VC, Overstreet C, Deak JD, Grant SFA, Polimanti R, Roussos P, Sun YV, Venkatesh S, Voloudakis G, Justice A, Begoli E, Ramoni R, Tourassi G, Pyarajan S, Tsao PS, O’Donnell CJ, Muralidhar S, Moser J, Casas JP, Bick AG, Zhou W, Cai T, Voight BF, Cho K, Gaziano MJ, Madduri RK, Damrauer SM, Liao KP. Diversity and Scale: Genetic Architecture of 2,068 Traits in the VA Million Veteran Program. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.28.23291975. [PMID: 37425708 PMCID: PMC10327290 DOI: 10.1101/2023.06.28.23291975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Genome-wide association studies (GWAS) have underrepresented individuals from non-European populations, impeding progress in characterizing the genetic architecture and consequences of health and disease traits. To address this, we present a population-stratified phenome-wide GWAS followed by a multi-population meta-analysis for 2,068 traits derived from electronic health records of 635,969 participants in the Million Veteran Program (MVP), a longitudinal cohort study of diverse U.S. Veterans genetically similar to the respective African (121,177), Admixed American (59,048), East Asian (6,702), and European (449,042) superpopulations defined by the 1000 Genomes Project. We identified 38,270 independent variants associating with one or more traits at experiment-wide P < 4.6 × 10 - 11 significance; fine-mapping 6,318 signals identified from 613 traits to single-variant resolution. Among these, a third (2,069) of the associations were found only among participants genetically similar to non-European reference populations, demonstrating the importance of expanding diversity in genetic studies. Our work provides a comprehensive atlas of phenome-wide genetic associations for future studies dissecting the architecture of complex traits in diverse populations.
Collapse
Affiliation(s)
- Anurag Verma
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
- Palo Alto Veterans Institute for Research (PAVIR), Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Alex Rodriguez
- Data Science and Learning, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Mitchell Conery
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Molei Liu
- Department of Biostatistics, Columbia University’s Mailman School of Public Health, New York, NY, 10032, USA
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Youngdae Kim
- Mathematics and Computer Science Division, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - David A Heise
- National Security Sciences Directorate, Cyber Resilience and Intelligence Division, Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Lindsay Guare
- Department of Medicine, Division of Translational Medicine and Human Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | | | - Helene Garcon
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Franciel Linares
- R&D Systems Engineering, Information Technology Services Directorate, Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Lauren Costa
- MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, 02111, USA
| | - Ian Goethert
- Data Management and Engineering, Information Technology Services Division, Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Ryan Tipton
- Knowledge Discovery Infrastructure, Information Technology Services Division, Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Jacqueline Honerlaw
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Laura Davies
- Computing and Computational Sciences Dir PMO, PMO, Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Stacey Whitbourne
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, 02111, USA
- Department of Medicine, Division of Aging, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - Jeremy Cohen
- National Security Sciences Directorate, Cyber Resilience and Intelligence Division, Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Daniel C Posner
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Rahul Sangar
- MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, 02111, USA
| | - Michael Murray
- MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, 02111, USA
| | - Xuan Wang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Daniel R Dochtermann
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Poornima Devineni
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Yunling Shi
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Tarak Nath Nandi
- Data Science and Learning, Argonne National Laboratory, Lemont, IL, 60439, USA
| | | | - Charles A Brunette
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Research Service, VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Robert J Carroll
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37211, USA
| | - Royce Clifford
- Research Department, VA San Diego Healthcare System, San Diego, CA, 92161, USA
- Surgery, Otolaryngology, UCSD San Diego, La Jolla, California, 92093, USA
| | - Scott Duvall
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, 84148, USA
- Internal Medicine, Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Joel Gelernter
- Psychiatry, Human Genetics, Yale University, New Haven, CT, 06520, USA
- VA Connecticut Healthcare System West Haven, West Haven, CT, 06516, USA
| | - Adriana Hung
- Medicine, Nephrology & Hypertension, VA Tennessee Valley Healthcare System & Vanderbilt University, Nashville, TN, 37232, USA
| | - Sudha K Iyengar
- Population and Quantitative Health Sciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - Jacob Joseph
- Medicine, Cardiology Section, VA Providence Healthcare System, Providence, RI, 02908, USA
- Department of Medicine, Brown University, Providence, RI, 02908, USA
| | - Rachel Kember
- Mental Illness Research, Education and Clinical Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Psychiatry, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Henry Kranzler
- Mental Illness Research, Education and Clinical Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Psychiatry, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Daniel Levey
- Psychiatry, Human Genetics, Yale University, New Haven, CT, 06520, USA
- Medicine, VA Connecticut Healthcare System West Haven, West Haven, CT, 06516, USA
| | - Shiuh-Wen Luoh
- VA Portland Health Care System, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Victoria C Merritt
- Research Department, VA San Diego Healthcare System, San Diego, CA, 92161, USA
| | - Cassie Overstreet
- Psychiatry, Human Genetics, Yale University, New Haven, CT, 06520, USA
| | - Joseph D Deak
- Psychiatry, Yale University, New Haven, CT, 06520, USA
- Psychiatry, VA Connecticut Healthcare System West Haven, West Haven, CT, 06516, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Divisions of Human Genetics and Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | | | - Panos Roussos
- Psychiatry, Mental Illness Research, Education and Clinical Center, James J. Peters VA Medical Center; Icahn School of Medicine at Mount Sinai, Bronx, NY, 10468, USA
| | - Yan V Sun
- Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, 30322, USA
| | - Sanan Venkatesh
- Psychiatry, Mental Illness Research, Education and Clinical Center, James J. Peters VA Medical Center; Icahn School of Medicine at Mount Sinai, Bronx, NY, 10468, USA
| | - Georgios Voloudakis
- Psychiatry, Mental Illness Research, Education and Clinical Center, James J. Peters VA Medical Center; Icahn School of Medicine at Mount Sinai, Bronx, NY, 10468, USA
| | - Amy Justice
- Medicine, VA Connecticut Healthcare System West Haven, West Haven, CT, 06516, USA
- Internal Medicine, General Medicine, Yale University, New Haven, CT, 06520, USA
- Health Policy, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Edmon Begoli
- Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Rachel Ramoni
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, 20420, USA
| | - Georgia Tourassi
- National Center for Computational Sciences, Oak Ridge National Laboratory, Dept of Energy, Oak Ridge, TN, 37831, USA
| | - Saiju Pyarajan
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Philip S Tsao
- Medicine, Cardiology, VA Palo Alto Healthcare System, Palo Alto, CA, 94304, USA
- Department of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | | | - Sumitra Muralidhar
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, 20420, USA
| | - Jennifer Moser
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, 20420, USA
| | - Juan P Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Alexander G Bick
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University, Nashville, TN, 37325, USA
| | - Wei Zhou
- Department of Medicine, Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- Stanley Center for Psychiatric Research, Cambridge, MA, 02142, USA
- Program in Medical and Population Genetics, Cambridge, MA, 02142, USA
| | - Tianxi Cai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Benjamin F Voight
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Kelly Cho
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, 02111, USA
- Department of Medicine, Division of Aging, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - Michael J Gaziano
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- MVP Boston Coordinating Center, VA Boston Healthcare System, Boston, MA, 02111, USA
- Department of Medicine, Division of Aging, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - Ravi K Madduri
- Data Science and Learning, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Scott M Damrauer
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Surgery, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Cardiovascular Institute, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Katherine P Liao
- Medicine, Rheumatology, VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| |
Collapse
|
137
|
Carracedo M, Ericson E, Ågren R, Forslöw A, Madeyski-Bengtson K, Svensson A, Riddle R, Christoffersson J, González-King Garibotti H, Lazovic B, Hicks R, Buvall L, Fornoni A, Greasley PJ, Lal M. APOL1 promotes endothelial cell activation beyond the glomerulus. iScience 2023; 26:106830. [PMID: 37250770 PMCID: PMC10209455 DOI: 10.1016/j.isci.2023.106830] [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: 03/30/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Apolipoprotein L1 (APOL1) high-risk genotypes are associated with increased risk of chronic kidney disease (CKD) in people of West African ancestry. Given the importance of endothelial cells (ECs) in CKD, we hypothesized that APOL1 high-risk genotypes may contribute to disease via EC-intrinsic activation and dysfunction. Single cell RNA sequencing (scRNA-seq) analysis of the Kidney Precision Medicine Project dataset revealed APOL1 expression in ECs from various renal vascular compartments. Utilizing two public transcriptomic datasets of kidney tissue from African Americans with CKD and a dataset of APOL1-expressing transgenic mice, we identified an EC activation signature; specifically, increased intercellular adhesion molecule 1 (ICAM-1) expression and enrichment in leukocyte migration pathways. In vitro, APOL1 expression in ECs derived from genetically modified human induced pluripotent stem cells and glomerular ECs triggered changes in ICAM-1 and platelet endothelial cell adhesion molecule 1 (PECAM-1) leading to an increase in monocyte attachment. Overall, our data suggest the involvement of APOL1 as an inducer of EC activation in multiple renal vascular beds with potential effects beyond the glomerular vasculature.
Collapse
Affiliation(s)
- Miguel Carracedo
- Bioscience Renal, Research and Early Development, Cardiovascular , Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Elke Ericson
- Genome Engineering, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Ågren
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Forslöw
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Katja Madeyski-Bengtson
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Svensson
- Translational Genomics, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rebecca Riddle
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Jonas Christoffersson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Hernán González-King Garibotti
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bojana Lazovic
- Genome Engineering, Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | - Lisa Buvall
- Bioscience Renal, Research and Early Development, Cardiovascular , Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Peter J. Greasley
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Mark Lal
- Bioscience Renal, Research and Early Development, Cardiovascular , Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
138
|
Adeva-Andany MM, Funcasta-Calderón R, Fernández-Fernández C, Ameneiros-Rodríguez E, Vila-Altesor M, Castro-Quintela E. The metabolic effects of APOL1 in humans. Pflugers Arch 2023:10.1007/s00424-023-02821-z. [PMID: 37261508 PMCID: PMC10233197 DOI: 10.1007/s00424-023-02821-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/04/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
Harboring apolipoprotein L1 (APOL1) variants coded by the G1 or G2 alleles of the APOL1 gene increases the risk for collapsing glomerulopathy, focal segmental glomerulosclerosis, albuminuria, chronic kidney disease, and accelerated kidney function decline towards end-stage kidney disease. However, most subjects carrying APOL1 variants do not develop the kidney phenotype unless a second clinical condition adds to the genotype, indicating that modifying factors modulate the genotype-phenotype correlation. Subjects with an APOL1 high-risk genotype are more likely to develop essential hypertension or obesity, suggesting that carriers of APOL1 risk variants experience more pronounced insulin resistance compared to noncarriers. Likewise, arterionephrosclerosis (the pathological correlate of hypertension-associated nephropathy) and glomerulomegaly take place among carriers of APOL1 risk variants, and these pathological changes are also present in conditions associated with insulin resistance, such as essential hypertension, aging, and diabetes. Insulin resistance may contribute to the clinical features associated with the APOL1 high-risk genotype. Unlike carriers of wild-type APOL1, bearers of APOL1 variants show impaired formation of lipid droplets, which may contribute to inducing insulin resistance. Nascent lipid droplets normally detach from the endoplasmic reticulum into the cytoplasm, although the proteins that enable this process remain to be fully defined. Wild-type APOL1 is located in the lipid droplet, whereas mutated APOL1 remains sited at the endoplasmic reticulum, suggesting that normal APOL1 may participate in lipid droplet biogenesis. The defective formation of lipid droplets is associated with insulin resistance, which in turn may modulate the clinical phenotype present in carriers of APOL1 risk variants.
Collapse
Affiliation(s)
- María M Adeva-Andany
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain.
| | - Raquel Funcasta-Calderón
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Carlos Fernández-Fernández
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Eva Ameneiros-Rodríguez
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Matilde Vila-Altesor
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Elvira Castro-Quintela
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| |
Collapse
|
139
|
Pavanello C, Ossoli A. HDL and chronic kidney disease. ATHEROSCLEROSIS PLUS 2023; 52:9-17. [PMID: 37193017 PMCID: PMC10182177 DOI: 10.1016/j.athplu.2023.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/18/2023]
Abstract
Low HDL-cholesterol (HDL-C) concentrations are a typical trait of the dyslipidemia associated with chronic kidney disease (CKD). In this condition, plasma HDLs are characterized by alterations in structure and function, and these particles can lose their atheroprotective functions, e.g., the ability to promote cholesterol efflux from peripheral cells, anti-oxidant and anti-inflammatory proprieties and they can even become dysfunctional, i.e., exactly damaging. The reduction in plasma HDL-C levels appears to be the only lipid alteration clearly linked to the progression of renal disease in CKD patients. The association between the HDL system and CKD development and progression is also supported by the presence of genetic kidney alterations linked to HDL metabolism, including mutations in the APOA1, APOE, APOL and LCAT genes. Among these, renal disease associated with LCAT deficiency is well characterized and lipid abnormalities detected in LCAT deficiency carriers mirror the ones observed in CKD patients, being present also in acquired LCAT deficiency. This review summarizes the major alterations in HDL structure and function in CKD and how genetic alterations in HDL metabolism can be linked to kidney dysfunction. Finally, the possibility of targeting the HDL system as possible strategy to slow CKD progression is reviewed.
Collapse
Affiliation(s)
| | - Alice Ossoli
- Corresponding author. Center E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133, Milano, Italy.
| |
Collapse
|
140
|
Acharya R, Upadhyay K. Early recurrence of focal segmental glomerulosclerosis in a kidney transplant recipient with APOL1 one risk variant. BMJ Case Rep 2023; 16:e254593. [PMID: 37258049 PMCID: PMC10254708 DOI: 10.1136/bcr-2023-254593] [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: 06/02/2023] Open
Abstract
Apolipoprotein 1 (APOL1) risk variants (G1 and G2) are associated with focal segmental glomerulosclerosis (FSGS) in patients of African ancestry. The prevalence of APOL1 two risk variants is lower in Hispanics and very rare in European and Asian populations. APOL1 two risk variants in donor kidneys is associated with recipient kidney graft loss, however the effect of recipient risk variant in the kidney transplant outcome is unclear. Here, we present a late adolescent male with FSGS and end stage renal disease with one APOL1 risk variant (G2) who had immediate recurrence of FSGS in the post-KT period. There was an excellent response to few sessions of plasmapheresis and Rituximab with no further recurrence of FSGS in the 1 year follow-up period. It needs to be seen whether the recipient APOL1 single risk variant causes increased susceptibility to kidney graft loss on a long run via recurrent or de novo pathologies.
Collapse
Affiliation(s)
- Ratna Acharya
- Paediatrics, University of Florida, Gainesville, Florida, USA
| | - Kiran Upadhyay
- Paediatrics and Nephrology, University of Florida Health, Gainesville, Florida, USA
| |
Collapse
|
141
|
Sandholm N, Dahlström EH, Groop PH. Genetic and epigenetic background of diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1163001. [PMID: 37324271 PMCID: PMC10262849 DOI: 10.3389/fendo.2023.1163001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Diabetic kidney disease (DKD) is a severe diabetic complication that affects up to half of the individuals with diabetes. Elevated blood glucose levels are a key underlying cause of DKD, but DKD is a complex multifactorial disease, which takes years to develop. Family studies have shown that inherited factors also contribute to the risk of the disease. During the last decade, genome-wide association studies (GWASs) have emerged as a powerful tool to identify genetic risk factors for DKD. In recent years, the GWASs have acquired larger number of participants, leading to increased statistical power to detect more genetic risk factors. In addition, whole-exome and whole-genome sequencing studies are emerging, aiming to identify rare genetic risk factors for DKD, as well as epigenome-wide association studies, investigating DNA methylation in relation to DKD. This article aims to review the identified genetic and epigenetic risk factors for DKD.
Collapse
Affiliation(s)
- Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emma H. Dahlström
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
142
|
Smith JD, Agrawal A, Wicklund C, Duquette D, Friedewald J, Rasmussen LV, Gacki-Smith J, Tandon SD, Muhammad LN, Yancy CW, Dong S, Cooper M, Gilbert A, Shetty A, Gordon EJ. Implementation of a culturally competent APOL1 genetic testing programme into living donor evaluation: A two-site, non-randomised, pre-post trial design. BMJ Open 2023; 13:e067657. [PMID: 37188469 PMCID: PMC10186444 DOI: 10.1136/bmjopen-2022-067657] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
INTRODUCTION While living donor (LD) kidney transplantation is the optimal treatment for patients with kidney failure, LDs assume a higher risk of future kidney failure themselves. LDs of African ancestry have an even greater risk of kidney failure post-donation than White LDs. Because evidence suggests that Apolipoprotein L1 (APOL1) risk variants contribute to this greater risk, transplant nephrologists are increasingly using APOL1 genetic testing to evaluate LD candidates of African ancestry. However, nephrologists do not consistently perform genetic counselling with LD candidates about APOL1 due to a lack of knowledge and skill in counselling. Without proper counselling, APOL1 testing will magnify LD candidates' decisional conflict about donating, jeopardising their informed consent. Given cultural concerns about genetic testing among people of African ancestry, protecting LD candidates' safety is essential to improve informed decisions about donating. Clinical 'chatbots', mobile apps that provide genetic information to patients, can improve informed treatment decisions. No chatbot on APOL1 is available and no nephrologist training programmes are available to provide culturally competent counselling to LDs about APOL1. Given the shortage of genetic counsellors, increasing nephrologists' genetic literacy is critical to integrating genetic testing into practice. METHODS AND ANALYSIS Using a non-randomised, pre-post trial design in two transplant centres (Chicago, IL, and Washington, DC), we will evaluate the effectiveness of culturally competent APOL1 testing, chatbot and counselling on LD candidates' decisional conflict about donating, preparedness for decision-making, willingness to donate and satisfaction with informed consent and longitudinally evaluate the implementation of this intervention into clinical practice using the Reach, Effectiveness, Adoption, Implementation and Maintenance framework. ETHICS AND DISSEMINATION This study will create a model for APOL1 testing of LDs of African ancestry, which can be implemented nationally via implementation science approaches. APOL1 will serve as a model for integrating culturally competent genetic testing into transplant and other practices to improve informed consent. This study involves human participants and was approved by Northwestern University IRB (STU00214038). Participants gave informed consent to participate in the study before taking part. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04910867. Registered 8 May 2021, https://register. CLINICALTRIALS gov/prs/app/action/SelectProtocol?sid=S000AWZ6&selectaction=Edit&uid=U0001PPF&ts=7&cx=-8jv7m2 ClinicalTrials.gov Identifier: NCT04999436. Registered 5 November 2021, https://register. CLINICALTRIALS gov/prs/app/action/SelectProtocol?sid=S000AYWW&selectaction=Edit&uid=U0001PPF&ts=11&cx=9tny7v.
Collapse
Affiliation(s)
- Justin D Smith
- Department of Population Health Sciences, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah, USA
- Departments of Psychiatry and Behavioral Sciences and Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Akansha Agrawal
- Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Catherine Wicklund
- Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Debra Duquette
- Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - John Friedewald
- Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Luke V Rasmussen
- Division of Health and Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jessica Gacki-Smith
- Center for Health Services and Outcomes Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - S Darius Tandon
- Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lutfiyya N Muhammad
- Department of Preventive Medicine-Division of Biostatistics, Northwestern University, Chicago, Illinois, USA
| | - Clyde W Yancy
- Department of Medicine-Division of Cardiology, Northwestern University, Evanston, Illinois, USA
| | - Siyuan Dong
- Department of Preventive Medicine-Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew Cooper
- Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alexander Gilbert
- Medicine, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Aneesha Shetty
- Medicine, The University of Arizona College of Medicine Tucson, Tucson, Arizona, USA
| | - Elisa J Gordon
- Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
143
|
Medina E, Rueda C, Batlle D. FSGS and COVID-19 in Non-African American Patients. KIDNEY360 2023; 4:687-699. [PMID: 37229730 PMCID: PMC10371264 DOI: 10.34067/kid.0000000000000104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/10/2023] [Indexed: 05/27/2023]
Abstract
Collapsing Focal Segmental Glomerulosclerosis (FSGS) has been reported relatively frequently in African American (AA) patients with coronavirus disease 2019 (COVID-19), and it is associated almost always with Apolipoprotein L gen 1 (APOL1) high-risk variants. We reviewed the published literature from April 2020 to November 2022 searching for non-African American (non-AA) patients with FSGS associated with COVID-19 (eight White patients, six Hispanic patients, three Asian patients, one Indian patient, and one Asian Indian patient). The following histologic patterns were found: collapsing (n=11), not otherwise specified (n=5), tip (n=2), and perihilar (n=1). Fifteen of the 19 patients had AKI. The APOL1 genotype was reported in only six of the 19 non-AA patients. Three of them (two Hispanic patients and one White patient) with collapsing FSGS had high-risk APOL1 variants. The other three patients (two White patients and one Hispanic patient with the collapsing variant, tip variant, and not otherwise specified) had low-risk APOL1 variants. Among 53 African American patients with collapsing FSGS associated with COVID-19, 48 had high-risk APOL1 variants and five had low-risk APOL1 variants. We conclude that in non-AA patients, FSGS is a rare complication of COVID-19. FSGS associated with COVID-19 can occur rarely with low-risk APOL1 variants in non-AA and AA patients. Non-AA patients reported to be associated with high-risk APOL1 variants possibly reflect inaccuracy of self-reported race with AA admixture because of unknown ancestry. Given the importance of APOL1 in the pathogenesis of FSGS associated with viral infection and to avoid racial bias, it seems appropriate that APOL1 testing be considered in patients with FSGS associated with COVID-19, regardless of self-reported race.
Collapse
Affiliation(s)
- Elba Medina
- Division of Nephrology, General Hospital of México, Eduardo Liceaga, México City, México
- Master's and PhD Program in Dental and Health Medical Sciences, Universidad Nacional Autónoma de México, México City, México
| | - Carlos Rueda
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Daniel Batlle
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
144
|
Smith KD, Akilesh S. Collapsing glomerulopathy: unraveling varied pathogeneses. Curr Opin Nephrol Hypertens 2023; 32:213-222. [PMID: 36811644 DOI: 10.1097/mnh.0000000000000873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
PURPOSE OF REVIEW Collapsing glomerulopathy presents clinically with nephrotic syndrome and rapid progressive loss of kidney function. Animal models and patient studies have uncovered numerous clinical and genetic conditions associated with collapsing glomerulopathy, as well as putative mechanisms, which will be reviewed here. RECENT FINDINGS Collapsing glomerulopathy is classified pathologically as a variant of focal and segmental glomerulosclerosis (FSGS). As such, most research efforts have focused on the causative role of podocyte injury in driving the disease. However, studies have also shown that injury to the glomerular endothelium or interruption of the podocyte-glomerular endothelial cell signaling axis can also cause collapsing glomerulopathy. Furthermore, emerging technologies are now enabling exploration of diverse molecular pathways that can precipitate collapsing glomerulopathy using biopsies from patients with the disease. SUMMARY Since its original description in the 1980s, collapsing glomerulopathy has been the subject of intense study, and these efforts have uncovered numerous insights into potential disease mechanisms. Newer technologies will enable profiling of the intra-patient and inter-patient variability in collapsing glomerulopathy mechanisms directly in patient biopsies, which will improve the diagnosis and classification of collapsing glomerulopathy.
Collapse
Affiliation(s)
- Kelly D Smith
- Department of Laboratory Medicine and Pathology, University of Washington
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington
- Kidney Research Institute, Seattle, Washington, USA
| |
Collapse
|
145
|
Elliott MD, Marasa M, Cocchi E, Vena N, Zhang JY, Khan A, Krishna Murthy S, Bheda S, Milo Rasouly H, Povysil G, Kiryluk K, Gharavi AG. Clinical and Genetic Characteristics of CKD Patients with High-Risk APOL1 Genotypes. J Am Soc Nephrol 2023; 34:909-919. [PMID: 36758113 PMCID: PMC10125632 DOI: 10.1681/asn.0000000000000094] [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: 10/25/2022] [Accepted: 01/04/2023] [Indexed: 02/11/2023] Open
Abstract
SIGNIFICANCE STATEMENT APOL1 high-risk genotypes confer a significant risk of kidney disease, but variability in patient outcomes suggests the presence of modifiers of the APOL1 effect. We show that a diverse population of CKD patients with high-risk APOL1 genotypes have an increased lifetime risk of kidney failure and higher eGFR decline rates, with a graded risk among specific high-risk genotypes. CKD patients with high-risk APOL1 genotypes have a lower diagnostic yield for monogenic kidney disease. Exome sequencing revealed enrichment of rare missense variants within the inflammasome pathway modifying the effect of APOL1 risk genotypes, which may explain some clinical heterogeneity. BACKGROUND APOL1 genotype has significant effects on kidney disease development and progression that vary among specific causes of kidney disease, suggesting the presence of effect modifiers. METHODS We assessed the risk of kidney failure and the eGFR decline rate in patients with CKD carrying high-risk ( N =239) and genetically matched low-risk ( N =1187) APOL1 genotypes. Exome sequencing revealed monogenic kidney diseases. Exome-wide association studies and gene-based and gene set-based collapsing analyses evaluated genetic modifiers of the effect of APOL1 genotype on CKD. RESULTS Compared with genetic ancestry-matched patients with CKD with low-risk APOL1 genotypes, those with high-risk APOL1 genotypes had a higher risk of kidney failure (Hazard Ratio [HR]=1.58), a higher decline in eGFR (6.55 versus 3.63 ml/min/1.73 m 2 /yr), and were younger at time of kidney failure (45.1 versus 53.6 years), with the G1/G1 genotype demonstrating the highest risk. The rate for monogenic kidney disorders was lower among patients with CKD with high-risk APOL1 genotypes (2.5%) compared with those with low-risk genotypes (6.7%). Gene set analysis identified an enrichment of rare missense variants in the inflammasome pathway in individuals with high-risk APOL1 genotypes and CKD (odds ratio=1.90). CONCLUSIONS In this genetically matched cohort, high-risk APOL1 genotypes were associated with an increased risk of kidney failure and eGFR decline rate, with a graded risk between specific high-risk genotypes and a lower rate of monogenic kidney disease. Rare missense variants in the inflammasome pathway may act as genetic modifiers of APOL1 effect on kidney disease.
Collapse
Affiliation(s)
- Mark D. Elliott
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Institute for Genomic Medicine, New York, NY
- Division of Nephrology, Department of Medicine, University of Calgary, Calgary, Canada
| | - Maddalena Marasa
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Enrico Cocchi
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Pediatrics, Universita’ degli Studi di Torino, Torino Italy
| | - Natalie Vena
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Institute for Genomic Medicine, New York, NY
| | - Jun Y. Zhang
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Sarath Krishna Murthy
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Shiraz Bheda
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Hila Milo Rasouly
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Gundula Povysil
- Columbia University Vagelos College of Physicians and Surgeons, Institute for Genomic Medicine, New York, NY
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Institute for Genomic Medicine, New York, NY
| | - Ali G. Gharavi
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Department of Medicine, Center for Precision Medicine and Genomics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, Institute for Genomic Medicine, New York, NY
| |
Collapse
|
146
|
Ilori TO, Brooks MS, Desai PN, Cheung KL, Judd SE, Crews DC, Cushman M, Winkler CA, Shlipak MG, Kopp JB, Naik RP, Estrella MM, Gutiérrez OM, Kramer H. Dietary Patterns, Apolipoprotein L1 Risk Genotypes, and CKD Outcomes Among Black Adults in the Reasons for Geographic and Racial Differences in Stroke (REGARDS) Cohort Study. Kidney Med 2023; 5:100621. [PMID: 37229446 PMCID: PMC10202773 DOI: 10.1016/j.xkme.2023.100621] [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] [Indexed: 03/08/2023] Open
Abstract
Rationale & Objective Dietary factors may impact inflammation and interferon production, which could influence phenotypic expression of Apolipoprotein1 (APOL1) genotypes. We investigated whether associations of dietary patterns with kidney outcomes differed by APOL1 genotypes. Study Design Prospective cohort. Settings & Participants 5,640 Black participants in the Reasons for Geographic and Racial Differences in Stroke (REGARDS). Exposures Five dietary patterns derived from food frequency questionnaires: Convenience foods, Southern, Sweets and Fats, Plant-based, and Alcohol/Salads. Outcomes Incident chronic kidney disease (CKD), CKD progression, and kidney failure. Incident CKD was defined as a change in estimated glomerular filtration rate (eGFR) to <60 mL/min/1.73 m2 accompanied by a ≥25% decline from baseline eGFR or development of kidney failure among those with baseline eGFR ≥60 mL/1.73 m2 body surface area. CKD progression was defined as a composite of 40% reduction in eGFR from baseline or development of kidney failure in the subset of participants who had serum creatinine levels at baseline and completed a second in-home visit/follow-up visit. Analytical Approach We examined associations of dietary pattern quartiles with incident CKD (n=4,188), CKD progression (n=5,640), and kidney failure (n=5,640). We tested for statistical interaction between dietary patterns and APOL1 genotypes for CKD outcomes and explored stratified analyses by APOL1 genotypes. Results Among 5,640 Black REGARDS participants, mean age was 64 years (standard deviation = 9), 35% were male, and 682 (12.1%) had high-risk APOL1 genotypes. Highest versus lowest quartiles (Q4 vs Q1) of Southern dietary pattern were associated with higher adjusted odds of CKD progression (OR, 1.28; 95% CI, 1.01-1.63) but not incident CKD (OR, 0.92; 95% CI, 0.74-1.14) or kidney failure (HR, 1.48; 95% CI, 0.90-2.44). No other dietary patterns showed significant associations with CKD. There were no statistically significant interactions between APOL1 genotypes and dietary patterns. Stratified analysis showed no consistent associations across genotypes, although Q3 and Q4 versus Q1 of Plant-based and Southern patterns were associated with lower odds of CKD progression among APOL1 high- but not low-risk genotypes. Limitations Included overlapping dietary patterns based on a single time point and multiple testing. Conclusions In Black REGARDS participants, Southern dietary pattern was associated with increased risk of CKD progression. Analyses stratified by APOL1 genotypes suggest associations may differ by genetic background, but these findings require confirmation in other cohorts.
Collapse
Affiliation(s)
- Titilayo O. Ilori
- Division of Nephrology, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA
| | - Marquita S. Brooks
- Department of Biostatistics, School of Public Health, University of Alabama, Birmingham, AB
| | - Parin N. Desai
- Division of Nephrology and Hypertension, Loyola University Chicago, Maywood, IL
| | - Katharine L. Cheung
- Division of Nephrology, Department of Medicine, Larner College of Medicine at The University of Vermont, Burlington, VT
| | - Suzanne E. Judd
- Department of Biostatistics, School of Public Health, University of Alabama, Birmingham, AB
| | - Deidra C. Crews
- Division of Nephrology, Department of Medicine, John Hopkins School of Medicine, Baltimore, MD
| | - Mary Cushman
- Division of Hematology, Department of Medicine, Larner College of Medicine at The University of Vermont, Burlington, VT
| | - Cheryl A. Winkler
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health and Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD
| | - Michael G. Shlipak
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD
| | - Rakhi P. Naik
- Division of Hematology, Department of Medicine, John Hopkins School of Medicine, Baltimore, MD
| | - Michelle M. Estrella
- Division of Nephrology, Department of Medicine, San Francisco VA Medical Center, San Francisco, CA
| | - Orlando M. Gutiérrez
- Division of Nephrology, Department of Medicine, University of Alabama, Birmingham, AB
| | - Holly Kramer
- Department of Public Health Sciences Division of Nephrology and Hypertension, Loyola University, Chicago, IL
| |
Collapse
|
147
|
Sanz AB, Sanchez-Niño MD, Ramos AM, Ortiz A. Regulated cell death pathways in kidney disease. Nat Rev Nephrol 2023; 19:281-299. [PMID: 36959481 PMCID: PMC10035496 DOI: 10.1038/s41581-023-00694-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 03/25/2023]
Abstract
Disorders of cell number that result from an imbalance between the death of parenchymal cells and the proliferation or recruitment of maladaptive cells contributes to the pathogenesis of kidney disease. Acute kidney injury can result from an acute loss of kidney epithelial cells. In chronic kidney disease, loss of kidney epithelial cells leads to glomerulosclerosis and tubular atrophy, whereas interstitial inflammation and fibrosis result from an excess of leukocytes and myofibroblasts. Other conditions, such as acquired cystic disease and kidney cancer, are characterized by excess numbers of cyst wall and malignant cells, respectively. Cell death modalities act to clear unwanted cells, but disproportionate responses can contribute to the detrimental loss of kidney cells. Indeed, pathways of regulated cell death - including apoptosis and necrosis - have emerged as central events in the pathogenesis of various kidney diseases that may be amenable to therapeutic intervention. Modes of regulated necrosis, such as ferroptosis, necroptosis and pyroptosis may cause kidney injury directly or through the recruitment of immune cells and stimulation of inflammatory responses. Importantly, multiple layers of interconnections exist between different modalities of regulated cell death, including shared triggers, molecular components and protective mechanisms.
Collapse
Affiliation(s)
- Ana B Sanz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Adrian M Ramos
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- RICORS2040, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain.
- RICORS2040, Madrid, Spain.
- Departamento de Farmacología, Universidad Autonoma de Madrid, Madrid, Spain.
| |
Collapse
|
148
|
Poppelaars F, Eskandari SK, Damman J, Seelen MA, Faria B, Gaya da Costa M. A non-muscle myosin heavy chain 9 genetic variant is associated with graft failure following kidney transplantation. Kidney Res Clin Pract 2023; 42:389-402. [PMID: 37313613 PMCID: PMC10265209 DOI: 10.23876/j.krcp.22.061] [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: 03/29/2022] [Revised: 06/06/2022] [Accepted: 07/07/2022] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Despite current matching efforts to identify optimal donor-recipient pairs for kidney transplantation, alloimmunity remains a major source of late transplant failure. Additional genetic parameters in donor-recipient matching could help improve longterm outcomes. Here, we studied the impact of a non-muscle myosin heavy chain 9 gene (MYH9) polymorphism on allograft failure. METHODS We conducted an observational cohort study, analyzing the DNA of 1,271 kidney donor-recipient transplant pairs from a single academic hospital for the MYH9 rs11089788 C>A polymorphism. The associations of the MYH9 genotype with risk of graft failure, biopsy-proven acute rejection (BPAR), and delayed graft function (DGF) were estimated. RESULTS A trend was seen in the association between the MYH9 polymorphism in the recipient and graft failure (recessive model, p = 0.056), but not for the MYH9 polymorphism in the donor. The AA-genotype MYH9 polymorphism in recipients was associated with higher risk of DGF (p = 0.03) and BPAR (p = 0.021), although significance was lost after adjusting for covariates (p = 0.15 and p = 0.10, respectively). The combined presence of the MYH9 polymorphism in donor-recipient pairs was associated with poor long-term kidney allograft survival (p = 0.04), in which recipients with an AA genotype receiving a graft with an AA genotype had the worst outcomes. After adjustment, this combined genotype remained significantly associated with 15-year death-censored kidney graft survival (hazard ratio, 1.68; 95% confidence interval, 1.05-2.70; p = 0.03). CONCLUSION Our results reveal that recipients with an AA-genotype MYH9 polymorphism receiving a donor kidney with an AA genotype have significantly elevated risk of graft failure after kidney transplantation.
Collapse
Affiliation(s)
- Felix Poppelaars
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Siawosh K. Eskandari
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jeffrey Damman
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marc A. Seelen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bernardo Faria
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Nephrology and Infectious Disease R&D Group, INEB, Institute of Investigation and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Mariana Gaya da Costa
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
149
|
Thomas CP, Daloul R, Lentine KL, Gohh R, Anand PM, Rasouly HM, Sharfuddin AA, Schlondorff JS, Rodig NM, Freese ME, Garg N, Lee BK, Caliskan Y. Genetic evaluation of living kidney donor candidates: A review and recommendations for best practices. Am J Transplant 2023; 23:597-607. [PMID: 36868514 DOI: 10.1016/j.ajt.2023.02.020] [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: 12/05/2022] [Revised: 01/25/2023] [Accepted: 02/20/2023] [Indexed: 03/05/2023]
Abstract
The growing accessibility and falling costs of genetic sequencing techniques has expanded the utilization of genetic testing in clinical practice. For living kidney donation, genetic evaluation has been increasingly used to identify genetic kidney disease in potential candidates, especially in those of younger ages. However, genetic testing on asymptomatic living kidney donors remains fraught with many challenges and uncertainties. Not all transplant practitioners are aware of the limitations of genetic testing, are comfortable with selecting testing methods, comprehending test results, or providing counsel, and many do not have access to a renal genetic counselor or a clinical geneticist. Although genetic testing can be a valuable tool in living kidney donor evaluation, its overall benefit in donor evaluation has not been demonstrated and it can also lead to confusion, inappropriate donor exclusion, or misleading reassurance. Until more published data become available, this practice resource should provide guidance for centers and transplant practitioners on the responsible use of genetic testing in the evaluation of living kidney donor candidates.
Collapse
Affiliation(s)
- Christie P Thomas
- Department of of Internal Medicine and Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA; VA Medical Center, Iowa City, Iowa, USA.
| | - Reem Daloul
- Division of Nephrology, Department of Internal Medicine, Allegheny General Hospital, Pittsburgh, Pennsylvania, USA
| | - Krista L Lentine
- Saint Louis University Transplant Center, SSM Health Saint Louis University Hospital, St. Louis, Missouri, USA
| | - Reginald Gohh
- Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Prince M Anand
- Mid-Carolinas Transplant Center, Medical University of South Carolina, Lancaster, South Carolina, USA
| | - Hila Milo Rasouly
- Center for Precision Medicine and Genomics, Department of Medicine, Columbia University, New York City, New York, USA
| | - Asif A Sharfuddin
- Division of Nephrology and Transplant, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Johannes S Schlondorff
- Department of Internal Medicine, Ohio State University Medical Center, Columbus, Ohio, USA
| | - Nancy M Rodig
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Margaret E Freese
- Department of of Internal Medicine and Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Neetika Garg
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Brian K Lee
- Kidney/Pancreas Transplant Center, Dell Seton Medical Center, University of Texas at Austin, Austin, Texas, USA
| | - Yasar Caliskan
- Saint Louis University Transplant Center, SSM Health Saint Louis University Hospital, St. Louis, Missouri, USA
| |
Collapse
|
150
|
Kopp JB, Yoshida T. Phenotypes of APOL1 High-Risk Status Subjects. J Am Soc Nephrol 2023; 34:735-736. [PMID: 37126670 PMCID: PMC10371258 DOI: 10.1681/asn.0000000000000123] [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] [Indexed: 05/03/2023] Open
Affiliation(s)
- Jeffrey B Kopp
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda, MD
| | | |
Collapse
|