1
|
Shaffer A, Meyerowitz EA. Clinical Manifestations of SARS-CoV-2 Infection in Immunocompetent Adults in the Era of Widespread Population Immunity and Omicron Sublineage Viruses. Infect Dis Clin North Am 2025; 39:233-251. [PMID: 40068975 DOI: 10.1016/j.idc.2025.02.002] [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: 05/10/2025]
Abstract
While most SARS-CoV-2 infections and reinfections in the era of widespread population immunity with omicron subsub-lineage variants are mild for immunocompetent individuals, any manifestation previously seen during the pandemic phase is still possible. COVID-19 may affect any organ system. Previous infections and prior vaccines protect against symptomatic future SARS-CoV-2 infections, though this protection wanes over time.
Collapse
Affiliation(s)
- Alexander Shaffer
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467, USA; Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eric A Meyerowitz
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467, USA; Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
2
|
Pollak MR, Friedman DJ. APOL1-associated kidney disease: modulators of the genotype-phenotype relationship. Curr Opin Nephrol Hypertens 2025; 34:191-198. [PMID: 40047214 DOI: 10.1097/mnh.0000000000001068] [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: 03/27/2025]
Abstract
PURPOSE OF REVIEW Apolipoprotein-L1 (APOL1) G1 and G2 risk variants, found in people of recent west sub-Saharan African ancestry, dramatically increase the likelihood of kidney disease, yet the incomplete penetrance an diverse clinical manifestations underscore the need to understand the molecular and environmental factors that modulate APOL1-mediated toxicity. RECENT FINDINGS Recent studies confirm that risk variants exert a toxic gain-of-function effect, exacerbated by inflammatory triggers such as HIV infection and COVID-19. Epigenetic mechanisms and microRNA pathways further modulate APOL1 expression, influencing disease penetrance. Multiple models have clarified how subcellular localization, signal peptide processing, and interactions with the endoplasmic reticulum may contribute to pathogenesis. Therapeutic advances include inhibitors targeting APOL1 ion channel activity and strategies that block key inflammatory signaling pathways. SUMMARY These findings highlight a multifaceted disease process driven by both the intrinsic toxic potential of APOL1 variants and numerous extrinsic triggers. Understanding this complex interplay will be pivotal for risk stratification and the development of precision therapies, potentially improving outcomes for populations disproportionately affected by APOL1-associated kidney disease.
Collapse
Affiliation(s)
- Martin R Pollak
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
3
|
Mahalingasivam V, Faucon AL, Sjölander A, Bosi A, González-Ortiz A, Lando S, Fu EL, Nitsch D, Bruchfeld A, Evans M, Wing K, Mansfield KE, Tomlinson L, Carrero JJ. Kidney Function Decline After COVID-19 Infection. JAMA Netw Open 2024; 7:e2450014. [PMID: 39724377 DOI: 10.1001/jamanetworkopen.2024.50014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2024] Open
Abstract
Importance COVID-19 infection has been associated with acute kidney injury. However, its possible association with long-term kidney function is not well understood. Objective To investigate whether kidney function decline accelerated after COVID-19 compared with after other respiratory tract infections. Design, Setting, and Participants This cohort study used linked data from the Stockholm Creatinine Measurements (SCREAM) Project between February 1, 2018, and January 1, 2022, in Stockholm, Sweden. All hospitalized and nonhospitalized adults in the database with at least 1 estimated glomerular filtration rate (eGFR) measurement in the 2 years prior to a COVID-19 positive test result or pneumonia diagnosis were selected. Statistical analyses were conducted between June 2023 and October 2024. Exposure COVID-19 and pneumonia (including influenza). Main Outcomes and Measures Mean annual change in eGFR after COVID-19 and after pneumonia was calculated with a linear regression model. Results The COVID-19 cohort comprised 134 565 individuals (74 819 females [55.6%]; median [IQR] age, 51 [37-64] years). The pneumonia cohort consisted of 35 987 individuals (19 359 females [53.8%]; median [IQR] age, 71 [56-81] years). The median (IQR) baseline eGFR was 94 (79-107) mL/min/1.73m2 for the COVID-19 cohort and 79 (61-92) mL/min/1.73m2 for the pneumonia cohort. After adjustment for covariates, both infections demonstrated accelerated annual eGFR decline, with greater magnitude of decline after COVID-19 (3.4% [95% CI, 3.2%-3.5%] after COVID-19; 2.3% [95% CI, 2.1%-2.5%] after pneumonia). This decline was more severe among individuals hospitalized for COVID-19 (5.4%; 95% CI, 5.2%-5.6%) but remained similar among those hospitalized for pneumonia. Conclusions and Relevance This cohort study found an association between COVID-19 and accelerated decline in kidney function, particularly after hospitalization, compared with pneumonia. People who were hospitalized for COVID-19 should receive closer monitoring of kidney function to ensure early diagnosis and optimized management of chronic kidney disease to effectively prevent complications and further decline.
Collapse
Affiliation(s)
- Viyaasan Mahalingasivam
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Nephrology and Transplantation, Barts Health National Health Service Trust, London, United Kingdom
| | - Anne-Laure Faucon
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Epidemiology, Institut National de la Santé et de la Recherche Médicale U1018, Paris-Saclay University, Villejuif, France
| | - Arvid Sjölander
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Alessandro Bosi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ailema González-Ortiz
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Translational Research Center, Instituto Nacional de Pediatria, Mexico City, Mexico
| | - Stefania Lando
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Edouard L Fu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dorothea Nitsch
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- UK Kidney Association, Bristol, United Kingdom
| | - Annette Bruchfeld
- Department of Health, Medicine and Caring Science, Linköping University, Linköping, Sweden
- Unit of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Evans
- Unit of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Unit of Renal Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Kevin Wing
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Public Health, School of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Kathryn E Mansfield
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Laurie Tomlinson
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Juan-Jesús Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Thareja SK, Yang X, Upama PB, Abdullah A, Torres SP, Cocroft LJ, Bubolz M, McGaughey K, Lou X, Kamaraju S, Ahamed SI, Madiraju P, Kwitek AE, Whittle J, Franco Z. Equitable community-based participatory research engagement with communities of color drives All of Us Wisconsin genomic research priorities. J Am Med Inform Assoc 2024; 31:2940-2951. [PMID: 39441983 PMCID: PMC11631115 DOI: 10.1093/jamia/ocae265] [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: 04/01/2024] [Revised: 09/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
OBJECTIVE The NIH All of Us Research Program aims to advance personalized medicine by not only linking patient records, surveys, and genomic data but also engaging with participants, particularly from groups traditionally underrepresented in biomedical research (UBR). This study details how the dialogue between scientists and community members, including many from communities of color, shaped local research priorities. MATERIALS AND METHODS We recruited area quantitative, basic, and clinical scientists as well as community members from our Community and Participant Advisory Boards with a predetermined interest in All of Us research as members of a Special Interest Group (SIG). An expert community engagement scientist facilitated 6 SIG meetings over the year, explicitly fostering openness and flexibility during conversations. We qualitatively analyzed discussions using a social movement framework tailored for community-based participatory research (CBPR) mobilization. RESULTS The SIG evolved through CBPR stages of emergence, coalescence, momentum, and maintenance/integration. Researchers prioritized community needs above personal academic interests while community members kept discussions focused on tangible return of value to communities. One key outcome includes SIG-driven shifts in programmatic and research priorities of the All of Us Research Program in Southeastern Wisconsin. One major challenge was building equitable conversations that balanced scientific rigor and community understanding. DISCUSSION Our approach allowed for a rich dialogue to emerge. Points of connection and disconnection between community members and scientists offered important guidance for emerging areas of genomic inquiry. CONCLUSION Our study presents a robust foundation for future efforts to engage diverse communities in CBPR, particularly on healthcare concerns affecting UBR communities.
Collapse
Affiliation(s)
- Suma K Thareja
- Medical College of Wisconsin, Milwaukee, WI 53226, United States
- All of Us Wisconsin, Milwaukee, WI 53226, United States
| | - Xin Yang
- All of Us Wisconsin, Milwaukee, WI 53226, United States
| | | | | | | | | | | | | | - Xuelin Lou
- Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Sailaja Kamaraju
- Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | | | | | - Anne E Kwitek
- Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Jeffrey Whittle
- Medical College of Wisconsin, Milwaukee, WI 53226, United States
- All of Us Wisconsin, Milwaukee, WI 53226, United States
- Clement J Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, United States
| | - Zeno Franco
- Medical College of Wisconsin, Milwaukee, WI 53226, United States
- All of Us Wisconsin, Milwaukee, WI 53226, United States
- Clement J Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, United States
| |
Collapse
|
5
|
Mayamba Nlandu Y, Tannor EK, Bamikefa TA, Rissassi Makulo JR. Kidney damage associated with COVID-19: from the acute to the chronic phase. Ren Fail 2024; 46:2316885. [PMID: 38561236 PMCID: PMC10986440 DOI: 10.1080/0886022x.2024.2316885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 04/04/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) infection is well established as a systemic disease including kidney damage. The entry point into the renal cell remains the angiotensin-converting enzyme 2 (ACE-2) receptor and the spectrum of renal lesions is broad, with a clear predominance of structural and functional tubular lesions. The most common form of glomerular injury is collapsing glomerulopathy (CG), which is strongly associated with apolipoprotein L1(APOL-1) risk variants. These acute lesions, which are secondary to the direct or indirect effects of SARS-CoV-2, can progress to chronicity and are specific to long COVID-19 in the absence of any other cause. Residual inflammation associated with SARS-CoV-2 infection, in addition to acute kidney injury (AKI) as a transitional state with or without severe histological lesions, may be responsible for greater kidney function decline in mild-to-moderate COVID-19. This review discusses the evidence for renal histological markers of chronicity in COVID-19 patients and triggers of low-grade inflammation that may explain the decline in kidney function in the post-COVID-19 period.
Collapse
Affiliation(s)
- Yannick Mayamba Nlandu
- Nephrology Unit, Kinshasa University Hospital, Kinshasa, Democratic Republic of the Congo
| | - Elliot Koranteng Tannor
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Directorate of Medicine, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | | | | |
Collapse
|
6
|
Siew ED, Hellwege JN, Hung AM, Birkelo BC, Vincz AJ, Parr SK, Denton J, Greevy RA, Robinson-Cohen C, Liu H, Susztak K, Matheny ME, Velez Edwards DR. Genome-wide association study of hospitalized patients and acute kidney injury. Kidney Int 2024; 106:291-301. [PMID: 38797326 PMCID: PMC11260539 DOI: 10.1016/j.kint.2024.04.019] [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: 06/29/2023] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 05/29/2024]
Abstract
Acute kidney injury (AKI) is a common and devastating complication of hospitalization. Here, we identified genetic loci associated with AKI in patients hospitalized between 2002-2019 in the Million Veteran Program and data from Vanderbilt University Medical Center's BioVU. AKI was defined as meeting a modified KDIGO Stage 1 or more for two or more consecutive days or kidney replacement therapy. Control individuals were required to have one or more qualifying hospitalizations without AKI and no evidence of AKI during any other observed hospitalizations. Genome-wide association studies (GWAS), stratified by race, adjusting for sex, age, baseline estimated glomerular filtration rate (eGFR), and the top ten principal components of ancestry were conducted. Results were meta-analyzed using fixed effects models. In total, there were 54,488 patients with AKI and 138,051 non-AKI individuals included in the study. Two novel loci reached genome-wide significance in the meta-analysis: rs11642015 near the FTO locus on chromosome 16 (obesity traits) (odds ratio 1.07 (95% confidence interval, 1.05-1.09)) and rs4859682 near the SHROOM3 locus on chromosome 4 (glomerular filtration barrier integrity) (odds ratio 0.95 (95% confidence interval, 0.93-0.96)). These loci colocalized with previous studies of kidney function, and genetic correlation indicated significant shared genetic architecture between AKI and eGFR. Notably, the association at the FTO locus was attenuated after adjustment for BMI and diabetes, suggesting that this association may be partially driven by obesity. Both FTO and the SHROOM3 loci showed nominal evidence of replication from diagnostic-code-based summary statistics from UK Biobank, FinnGen, and Biobank Japan. Thus, our large GWA meta-analysis found two loci significantly associated with AKI suggesting genetics may explain some risk for AKI.
Collapse
Affiliation(s)
- Edward D Siew
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for AKI Research (VIP-AKI), Nashville, Tennessee, USA.
| | - Jacklyn N Hellwege
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Adriana M Hung
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for AKI Research (VIP-AKI), Nashville, Tennessee, USA
| | - Bethany C Birkelo
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for AKI Research (VIP-AKI), Nashville, Tennessee, USA
| | - Andrew J Vincz
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for AKI Research (VIP-AKI), Nashville, Tennessee, USA
| | - Sharidan K Parr
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for AKI Research (VIP-AKI), Nashville, Tennessee, USA
| | - Jason Denton
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA
| | - Robert A Greevy
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cassianne Robinson-Cohen
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease (VCKD) and Integrated Program for AKI Research (VIP-AKI), Nashville, Tennessee, USA
| | - Hongbo Liu
- Division of Renal Electrolyte and Hypertension, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA; Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Division of Renal Electrolyte and Hypertension, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA; Philadelphia Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Michael E Matheny
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Digna R Velez Edwards
- Tennessee Valley Health Systems, Nashville Veterans Affairs, Nashville, Tennessee, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
7
|
Himmelfarb J, Stanaway IB, Bhatraju PK. Acute kidney injury genetic risks: taking it 1 SNP at a time. Kidney Int 2024; 106:188-190. [PMID: 39032964 DOI: 10.1016/j.kint.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/08/2024] [Indexed: 07/23/2024]
Abstract
This commentary addresses some of the strengths, shortcomings, and challenges of the genome-wide association study of acute kidney injury (AKI) report in this issue. This AKI genome-wide association study is well executed and provides significant progress in finding 2 genome-wide significant loci. However, significant interpretive challenges remain, where advancements in methods are needed because of the clinical heterogeneity of the AKI phenotype, plus possible bias due to genetic correlation between index hospitalization risk and AKI risk.
Collapse
Affiliation(s)
- Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ian B Stanaway
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Pavan K Bhatraju
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA; School of Medicine, Sepsis Center of Research Excellence, University of Washington, Seattle, Washington, USA
| |
Collapse
|
8
|
Babroudi S, Weiner DE, Neyra JA, Drew DA. Acute Kidney Injury Receiving Dialysis and Dialysis Care after Hospital Discharge. J Am Soc Nephrol 2024; 35:962-971. [PMID: 38652567 PMCID: PMC11230726 DOI: 10.1681/asn.0000000000000383] [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: 11/29/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
The number of patients with AKI receiving outpatient hemodialysis (AKI-D) is increasing. At present, on the basis of limited data, approximately one third of patients with AKI-D who receive outpatient dialysis after hospital discharge survive and regain sufficient kidney function to discontinue dialysis. Data to inform dialysis management strategies that promote kidney function recovery and processes of care among patients with AKI-D receiving outpatient dialysis are lacking. In this article, we detail current trends in the incidence, risk factors, clinical outcomes, proposed management, and health policy landscape for patients with AKI-D receiving outpatient dialysis and identify areas for further research.
Collapse
Affiliation(s)
- Seda Babroudi
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Daniel E. Weiner
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Javier A. Neyra
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David A. Drew
- Division of Nephrology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
9
|
Tabachnikov O, Skorecki K, Kruzel-Davila E. APOL1 nephropathy - a population genetics success story. Curr Opin Nephrol Hypertens 2024; 33:447-455. [PMID: 38415700 PMCID: PMC11139250 DOI: 10.1097/mnh.0000000000000977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
PURPOSE OF REVIEW More than a decade ago, apolipoprotein L1 ( APOL1 ) risk alleles designated G1 and G2, were discovered to be causally associated with markedly increased risk for progressive kidney disease in individuals of recent African ancestry. Gratifying progress has been made during the intervening years, extending to the development and clinical testing of genomically precise small molecule therapy accompanied by emergence of RNA medicine platforms and clinical testing within just over a decade. RECENT FINDINGS Given the plethora of excellent prior review articles, we will focus on new findings regarding unresolved questions relating mechanism of cell injury with mode of inheritance, regulation and modulation of APOL1 activity, modifiers and triggers for APOL1 kidney risk penetrance, the pleiotropic spectrum of APOL1 related disease beyond the kidney - all within the context of relevance to therapeutic advances. SUMMARY Notwithstanding remaining controversies and uncertainties, promising genomically precise therapies targeted at APOL1 mRNA using antisense oligonucleotides (ASO), inhibitors of APOL1 expression, and small molecules that specifically bind and inhibit APOL1 cation flux are emerging, many already at the clinical trial stage. These therapies hold great promise for mitigating APOL1 kidney injury and possibly other systemic phenotypes as well. A challenge will be to develop guidelines for appropriate use in susceptible individuals who will derive the greatest benefit.
Collapse
Affiliation(s)
- Orly Tabachnikov
- Department of Nephrology, Rambam Healthcare Campus, Haifa, Israel
| | - Karl Skorecki
- Department of Nephrology, Rambam Healthcare Campus, Haifa, Israel
- Departments of Genetics and Developmental Biology and Rappaport Faculty of Medicine and Research Institute, Technion—Israel Institute of Technology, Haifa, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Etty Kruzel-Davila
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Department of Nephrology, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
10
|
Itoku A, Isaac J, Wilson S, Reidy K, Kaskel F. APOL1 Nephropathy Risk Variants Through the Life Course: A Review. Am J Kidney Dis 2024; 84:102-110. [PMID: 38341125 DOI: 10.1053/j.ajkd.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/26/2023] [Accepted: 12/05/2023] [Indexed: 02/12/2024]
Abstract
Two variant alleles of the gene apolipoprotein L1 (APOL1), known as risk variants (RVs), are a major contributor to kidney disease burden in those of African descent. The APOL1 protein contributes to innate immunity and may protect against Trypanosoma, HIV, Salmonella, and leishmaniasis. However, the effects of carrying 1 or more RVs contribute to a variety of disease processes starting as early as in utero and can be exacerbated by other factors (or "second hits"). Indeed, these genetic variations interact with environmental exposures, infections, and systemic disease to modify health outcomes across the life span. This review focuses on APOL1-associated diseases through the life-course perspective and discusses how early exposure to second hits can impact long-term outcomes. APOL1-related kidney disease typically presents in adolescents to young adults, and individuals harboring RVs are more likely to progress to kidney failure than are those with kidney disease who lack APOL-1 RVs. Ongoing research is aimed at elucidating the association of APOL1 RV effects with adverse donor and recipient kidney transplant outcomes. Unfortunately, there is currently no established treatment for APOL1-associated nephropathy. Long-term research is needed to evaluate the risk and protective factors associated with APOL1 RVs at different stages of life.
Collapse
Affiliation(s)
- Ai Itoku
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Jaya Isaac
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Scott Wilson
- Albert Einstein College of Medicine, Bronx, New York.
| | - Kimberly Reidy
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Frederick Kaskel
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| |
Collapse
|
11
|
Li Y, Gong Y, Xu G. New insights into kidney disease after COVID-19 infection and vaccination: histopathological and clinical findings. QJM 2024; 117:317-337. [PMID: 37402613 DOI: 10.1093/qjmed/hcad159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Indexed: 07/06/2023] Open
Abstract
In addition to its pulmonary effects, coronavirus disease 2019 (COVID-19) has also been found to cause acute kidney injury (AKI), which has been linked to high mortality rates. In this review, we collected data from 20 clinical studies on post-COVID-19-related AKI and 97 cases of AKI associated with COVID-19 vaccination. Acute tubular injury was by far the most common finding in the kidneys of patients with COVID-19-related AKI. Among patients hospitalized for COVID-19, 34.0% developed AKI, of which 59.0%, 19.1% and 21.9% were Stages 1, 2 and 3, respectively. Though kidney disease and other adverse effects after COVID-19 vaccination overall appear rare, case reports have accumulated suggesting that COVID-19 vaccination may be associated with a risk of subsequent kidney disease. Among the patients with post-vaccination AKI, the most common pathologic findings include crescentic glomerulonephritis (29.9%), acute tubular injury (23.7%), IgA nephropathy (18.6%), antineutrophil cytoplasmic autoantibody-associated vasculitis (17.5%), minimal change disease (17.5%) and thrombotic microangiopathy (10.3%). It is important to note that crescentic glomerulonephritis appears to be more prevalent in patients who have newly diagnosed renal involvement. The proportions of patients with AKI Stages 1, 2 and 3 after COVID-19 vaccination in case reports were 30.9%, 22.7% and 46.4%, respectively. In general, clinical cases of new-onset and recurrent nephropathy with AKI after COVID-19 vaccination have a positive prognosis. In this article, we also explore the underlying pathophysiological mechanisms of AKI associated with COVID-19 infection and its vaccination by describing key renal morphological and clinical features and prognostic findings.
Collapse
Affiliation(s)
- Yebei Li
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang 330006, P.R. China
| | - Yan Gong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang 330006, P.R. China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang 330006, P.R. China
| |
Collapse
|
12
|
Koehler S, Hengel FE, Dumoulin B, Damashek L, Holzman LB, Susztak K, Huber TB. The 14th International Podocyte Conference 2023: from podocyte biology to glomerular medicine. Kidney Int 2024; 105:935-952. [PMID: 38447880 DOI: 10.1016/j.kint.2024.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 03/08/2024]
Abstract
The 14th International Podocyte Conference took place in Philadelphia, Pennsylvania, USA from May 23 to 26, 2023. It commenced with an early-career researchers' meeting on May 23, providing young scientists with a platform to present and discuss their research findings. Throughout the main conference, 29 speakers across 9 sessions shared their insights on podocyte biology, glomerular medicine, novel technologic advancements, and translational approaches. Additionally, the event featured 3 keynote lectures addressing engineered chimeric antigen receptor T cell- and mRNA-based therapies and the use of biobanks for enhanced disease comprehension. Furthermore, 4 brief oral abstract sessions allowed scientists to present their findings to a broad audience. The program also included a panel discussion addressing the challenges of conducting human research within the American Black community. Remarkably, after a 5-year hiatus from in-person conferences, the 14th International Podocyte Conference successfully convened scientists from around the globe, fostering the presentation and discussion of crucial research findings, as summarized in this review. Furthermore, to ensure continuous and sustainable education, research, translation, and trial medicine related to podocyte and glomerular diseases for the benefit of patients, the International Society of Glomerular Disease was officially launched during the conference.
Collapse
Affiliation(s)
- Sybille Koehler
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Felicitas E Hengel
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Bernhard Dumoulin
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Laurel Damashek
- International Society of Glomerular Disease, Florence, Massachusetts, USA
| | - Lawrence B Holzman
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tobias B Huber
- III. Department of Medicine and Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; International Society of Glomerular Disease, Florence, Massachusetts, USA.
| |
Collapse
|
13
|
Adamson WE, Noyes H, Johnson P, Cooper A, Monckton DG, Ogunsola J, Beckett-Hill G, Sullivan M, Mark P, Parekh RS, MacLeod A. Phenome-wide analysis reveals epistatic associations between APOL1 variants and chronic kidney disease and multiple other disorders. EBioMedicine 2024; 101:105000. [PMID: 38360481 PMCID: PMC10944146 DOI: 10.1016/j.ebiom.2024.105000] [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: 06/26/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND APOL1 variants G1 and G2 are common in populations with recent African ancestry. They are associated with protection from African sleeping sickness, however homozygosity or compound heterozygosity for these variants is associated with chronic kidney disease (CKD) and related conditions. What is not clear is the extent of associations with non-kidney-related disorders, and whether there are clusters of diseases associated with individual APOL1 genotypes. METHODS Using a cohort of 7462 UK Biobank participants with recent African ancestry, we conducted a phenome-wide association study investigating associations between individual APOL1 genotypes and conditions identified by the International Classification of Disease phenotypes. FINDINGS We identified 27 potential associations between individual APOL1 genotypes and a diverse range of conditions. G1/G2 compound heterozygotes were specifically associated with 26 of these conditions (all deleteriously), with an over-representation of infectious diseases (including hospitalisation and death resulting from COVID-19). The analysis also exposed complexities in the relationship between APOL1 and CKD that are not evident when risk variants are grouped together: G1 homozygosity, G2 homozygosity, and G1/G2 compound heterozygosity were each shown to be associated with distinct CKD phenotypes. The multi-locus nature of the G1/G2 genotype means that its associations would go undetected in a standard genome-wide association study. INTERPRETATION Our findings have implications for understanding health risks and better-targeted detection, intervention, and therapeutic strategies, particularly in populations where APOL1 G1 and G2 are common such as in sub-Saharan Africa and its diaspora. FUNDING This study was funded by the Wellcome Trust (209511/Z/17/Z) and H3Africa (H3A/18/004).
Collapse
Affiliation(s)
- Walt E Adamson
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, United Kingdom; Wellcome Centre for Integrative Parasitology, University of Glasgow, United Kingdom; TrypanoGEN+ Research Group, Uganda, Member of the H3Africa Consortium, South Africa.
| | - Harry Noyes
- TrypanoGEN+ Research Group, Uganda, Member of the H3Africa Consortium, South Africa; Centre for Genomic Research, University of Liverpool, United Kingdom
| | - Paul Johnson
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, United Kingdom
| | - Anneli Cooper
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, United Kingdom; Wellcome Centre for Integrative Parasitology, University of Glasgow, United Kingdom
| | - Darren G Monckton
- School of Molecular Biosciences, University of Glasgow, United Kingdom
| | - John Ogunsola
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, United Kingdom; Wellcome Centre for Integrative Parasitology, University of Glasgow, United Kingdom
| | - Georgia Beckett-Hill
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, United Kingdom
| | - Michael Sullivan
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Patrick Mark
- School of Cardiovascular and Metabolic Health, University of Glasgow, United Kingdom
| | - Rulan S Parekh
- Women's College Hospital, Hospital for Sick Children and University of Toronto, Canada
| | - Annette MacLeod
- School of Biodiversity, One Health, and Veterinary Medicine, University of Glasgow, United Kingdom; Wellcome Centre for Integrative Parasitology, University of Glasgow, United Kingdom; TrypanoGEN+ Research Group, Uganda, Member of the H3Africa Consortium, South Africa.
| |
Collapse
|
14
|
Lukowsky LR, Der-Martirosian C, Northcraft H, Kalantar-Zadeh K, Goldfarb DS, Dobalian A. Predictors of Acute Kidney Injury (AKI) among COVID-19 Patients at the US Department of Veterans Affairs: The Important Role of COVID-19 Vaccinations. Vaccines (Basel) 2024; 12:146. [PMID: 38400130 PMCID: PMC10892207 DOI: 10.3390/vaccines12020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND There are knowledge gaps about factors associated with acute kidney injury (AKI) among COVID-19 patients. To examine AKI predictors among COVID-19 patients, a retrospective longitudinal cohort study was conducted between January 2020 and December 2022. Logistic regression models were used to examine predictors of AKI, and survival analysis was performed to examine mortality in COVID-19 patients. RESULTS A total of 742,799 veterans diagnosed with COVID-19 were included and 95,573 were hospitalized within 60 days following COVID-19 diagnosis. A total of 45,754 developed AKI and 28,573 AKI patients were hospitalized. Use of vasopressors (OR = 14.73; 95% CL 13.96-15.53), history of AKI (OR = 2.22; CL 2.15-2.29), male gender (OR = 1.90; CL 1.75-2.05), Black race (OR = 1.62; CL 1.57-1.65), and age 65+ (OR = 1.57; CL 1.50-1.63) were associated with AKI. Patients who were vaccinated twice and boosted were least likely to develop AKI (OR = 0.51; CL 0.49-0.53) compared to unvaccinated COVID-19 patients. Patients receiving two doses (OR = 0.77; CL = 0.72-0.81), or a single dose (OR = 0.88; CL = 0.81-0.95) were also less likely to develop AKI compared to the unvaccinated. AKI patients exhibited four times higher mortality compared to those without AKI (HR = 4.35; CL 4.23-4.50). Vaccinated and boosted patients had the lowest mortality risk compared to the unvaccinated (HR = 0.30; CL 0.28-0.31). CONCLUSION Use of vasopressors, being unvaccinated, older age, male gender, and Black race were associated with post COVID-19 AKI. Whether COVID-19 vaccination, including boosters, decreases the risk of developing AKI warrants additional studies.
Collapse
Affiliation(s)
- Lilia R. Lukowsky
- Veterans Emergency Management Evaluation Center (VEMEC), US Department of Veterans Affairs, North Hills, CA 91343, USA; (C.D.-M.); (H.N.); (A.D.)
| | - Claudia Der-Martirosian
- Veterans Emergency Management Evaluation Center (VEMEC), US Department of Veterans Affairs, North Hills, CA 91343, USA; (C.D.-M.); (H.N.); (A.D.)
| | - Heather Northcraft
- Veterans Emergency Management Evaluation Center (VEMEC), US Department of Veterans Affairs, North Hills, CA 91343, USA; (C.D.-M.); (H.N.); (A.D.)
| | - Kamyar Kalantar-Zadeh
- The Lundquist Institute for Biomedical Innovation, Harbor UCLA Medical Center, Torrance, CA 90502, USA;
- Tibor Rubin VA Medical Center, Long Beach VA Healthcare System, Long Beach, CA 90822, USA
| | - David S. Goldfarb
- New York Harbor VA Healthcare System (NYHHS), US Department of Veterans Affairs, New York, NY 10010, USA;
- NYU Langone Health, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Aram Dobalian
- Veterans Emergency Management Evaluation Center (VEMEC), US Department of Veterans Affairs, North Hills, CA 91343, USA; (C.D.-M.); (H.N.); (A.D.)
- Division of Health Services Management and Policy in the College of Public Health, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Bernal C, How-Volkman C, Spencer M, El-Shamy A, Mohieldin AM. The Role of Extracellular Vesicles in SARS-CoV-2-Induced Acute Kidney Injury: An Overview. Life (Basel) 2024; 14:163. [PMID: 38398672 PMCID: PMC10890680 DOI: 10.3390/life14020163] [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: 12/03/2023] [Revised: 01/09/2024] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected millions worldwide since its outbreak in the winter of 2019. While extensive research has primarily focused on the deleterious respiratory effects of SARS-CoV-2 in recent years, its pan-tropism has become evident. Among the vital organs susceptible to SARS-CoV-2 infection is the kidney. Post SARS-CoV-2 infection, patients have developed coronavirus disease 19 (COVID-19), with reported incidences of COVID-19 patients developing acute kidney injury (AKI). Given COVID-19's multisystemic manifestation, our review focuses on the impact of SARS-CoV-2 infection within the renal system with an emphasis on the current hypotheses regarding the role of extracellular vesicles (EVs) in SARS-CoV-2 pathogenesis. Emerging studies have shown that SARS-CoV-2 can directly infect the kidney, whereas EVs are involved in the spreading of SARS-CoV-2 particles to other neighboring cells. Once the viral particles are within the kidney system, many proinflammatory signaling pathways are shown to be activated, resulting in AKI. Hence, clinical investigation of urinary proinflammatory components and total urinary extracellular vesicles (uEVs) with viral particles have been used to assess the severity of AKI in patients with COVID-19. Remarkedly, new emerging studies have shown the potential of mesenchymal stem cell-derived EVs (MSC-EVs) and ACE2-containing EVs as a hopeful therapeutic tool to inhibit SARS-CoV-2 RNA replication and block viral entry, respectively. Overall, understanding EVs' physiological role is crucial and hopefully will rejuvenate our therapeutic approach towards COVID-19 patients with AKI.
Collapse
Affiliation(s)
- Carter Bernal
- College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Christiane How-Volkman
- College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Madison Spencer
- College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
| | - Ahmed El-Shamy
- College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Ashraf M. Mohieldin
- College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| |
Collapse
|
16
|
Hung AM, Assimon VA, Chen HC, Yu Z, Vlasschaert C, Triozzi JL, Chan H, Wheless L, Wilson O, Shah SC, Mack T, Thompson T, Matheny ME, Chandrasekar S, Mozaffari SV, Chung CP, Tsao P, Susztak K, Siew ED, Estrada K, Gaziano JM, Graham RR, Tao R, Hoek M, Robinson-Cohen C, Green EM, Bick AG. Genetic Inhibition of APOL1 Pore-Forming Function Prevents APOL1-Mediated Kidney Disease. J Am Soc Nephrol 2023; 34:1889-1899. [PMID: 37798822 PMCID: PMC10631602 DOI: 10.1681/asn.0000000000000219] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/17/2023] [Indexed: 10/07/2023] Open
Abstract
SIGNIFICANCE STATEMENT African Americans are at increased risk of CKD in part due to high-risk (HR) variants in the apolipoprotein L1 ( APOL1 ) gene, termed G1/G2. A different APOL1 variant, p.N264K , reduced the risk of CKD and ESKD among carriers of APOL1 HR variants to levels comparable with individuals with APOL1 low-risk variants in an analysis of 121,492 participants of African ancestry from the Million Veteran Program (MVP). Functional genetic studies in cell models showed that APOL1 p.N264K blocked APOL1 pore-forming function and ion channel conduction and reduced toxicity of APOL1 HR mutations. Pharmacologic inhibitors that mimic this mutation blocking APOL1 -mediated pore formation may be able to prevent and/or treat APOL1 -associated kidney disease. BACKGROUND African Americans are at increased risk for nondiabetic CKD in part due to HR variants in the APOL1 gene. METHODS We tested whether a different APOL1 variant, p.N264K , modified the association between APOL1 HR genotypes (two copies of G1/G2) and CKD in a cross-sectional analysis of 121,492 participants of African ancestry from the MVP. We replicated our findings in the Vanderbilt University Biobank ( n =14,386) and National Institutes of Health All of Us ( n =14,704). Primary outcome was CKD and secondary outcome was ESKD among nondiabetic patients. Primary analysis compared APOL1 HR genotypes with and without p.N264K . Secondary analyses included APOL1 low-risk genotypes and tested for interaction. In MVP, we performed sequential logistic regression models adjusting for demographics, comorbidities, medications, and ten principal components of ancestry. Functional genomic studies expressed APOL1 HR variants with and without APOL1 p.N264K in cell models. RESULTS In the MVP cohort, 15,604 (12.8%) had two APOL1 HR variants, of which 582 (0.5%) also had APOL1 p.N264K . In MVP, 18,831 (15%) had CKD, 4177 (3%) had ESKD, and 34% had diabetes. MVP APOL1 HR, without p.N264K , was associated with increased odds of CKD (odds ratio [OR], 1.72; 95% confidence interval [CI], 1.60 to 1.85) and ESKD (OR, 3.94; 95% CI, 3.52 to 4.41). In MVP, APOL1 p.N264K mitigated the renal risk of APOL1 HR, in CKD (OR, 0.43; 95% CI, 0.28 to 0.65) and ESKD (OR, 0.19; CI 0.07 to 0.51). In the replication cohorts meta-analysis, APOL1 p.N264K mitigated the renal risk of APOL1 HR in CKD (OR, 0.40; 95% CI, 0.18 to 0.92) and ESKD (OR, 0.19; 95% CI, 0.05 to 0.79). In the mechanistic studies, APOL1 p.N264K blocked APOL1 pore-forming function and ion channel conduction and reduced toxicity of APOL1 HR variants. CONCLUSIONS APOL1 p.N264K is associated with reduced risk of CKD and ESKD among carriers of APOL1 HR to levels comparable with individuals with APOL1 low-risk genotypes.
Collapse
Affiliation(s)
- Adriana M. Hung
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Hua-Chang Chen
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Zhihong Yu
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Jefferson L. Triozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Helen Chan
- Maze Therapeutics, South San Francisco, California
| | - Lee Wheless
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Otis Wilson
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shailja C. Shah
- VA San Diego Healthcare System and UC San Diego Health, La Jolla, California
| | - Taralynn Mack
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Trevor Thompson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael E. Matheny
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Cecilia P. Chung
- Department of Rheumatology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Philip Tsao
- VA Palo Alto Health Care System, Palo Alto, California
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Edward D. Siew
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - J. Michael Gaziano
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Brigham and Women's Hospital and Harvard School of Medicine, Boston, Massachusetts
| | | | - Ran Tao
- Nashville VA Medical Center, VA Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Maarten Hoek
- Maze Therapeutics, South San Francisco, California
| | - Cassianne Robinson-Cohen
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, Massachusetts
| |
Collapse
|
17
|
Whitbourne SB, Moser J, Cho K, Deen J, Churby LL, Justice AC, Casas JP, Pyarajan S, Tsao PS, Gaziano JM, Muralidhar S. Leveraging the Million Veteran Program Infrastructure and Data for a Rapid Research Response to COVID-19. Fed Pract 2023; 40:S23-S28. [PMID: 38577307 PMCID: PMC10988626 DOI: 10.12788/fp.0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Background The Veterans Health Administration Office of Research and Development (ORD) played a key role in the federal government's response to the COVID-19 pandemic. The ORD effectively leveraged existing resources to answer questions related to the SARS-CoV-2 virus and COVID-19. Observations When the COVID-19 pandemic hit in 2020, the Million Veteran Program (MVP), one of the largest genomic cohorts in the world, extended the centralized recruitment and enrollment infrastructure to develop a COVID-19 research volunteer registry to assist enrollment in the vaccine and treatment trials in which the US Department of Veterans Affairs (VA) participated. In addition, the MVP allowed for new data collection and a large genomic cohort to understand host contributions to COVID-19. This article describes ways the MVP contributed to the VA's rapid research response to COVID-19. Several host genetic factors believed to play a role in the development and severity of COVID-19 were identified. Furthermore, existing MVP partnerships with other federal agencies, particularly with the Department of Energy, were leveraged to improve understanding and management of COVID-19. Conclusions A previously established enterprise approach and research infrastructure were essential to the VA's successful and timely COVID-19 research response. This infrastructure not only supported rapid recruitment in vaccine and treatment trials, but also leveraged the unique MVP and VA electronic health record data to drive rapid scientific discovery and inform clinical operations. Extending the models that VA research applied to the federal government at large and establishing centralized resources for shared or federated data analyses across federal agencies will better equip the nation to respond to future public health crises.
Collapse
Affiliation(s)
- Stacey B. Whitbourne
- Veterans Affairs Boston Healthcare System, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Jennifer Moser
- Office of Research and Development, Department of Veterans Affairs, Washington, DC
| | - Kelly Cho
- Veterans Affairs Boston Healthcare System, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Jennifer Deen
- Office of Research and Development, Department of Veterans Affairs, Washington, DC
| | - Lori L. Churby
- Veterans Affairs Palo Alto Healthcare System, California
| | - Amy C. Justice
- Veterans Affairs Connecticut Healthcare System, West Haven
- Yale University School of Medicine and School of Public Health, New Haven, Connecticut
| | - Juan P. Casas
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts
| | - Saiju Pyarajan
- Veterans Affairs Boston Healthcare System, Massachusetts
| | - Phil S. Tsao
- Veterans Affairs Palo Alto Healthcare System, California
- Stanford University School of Medicine, Palo Alto, California
| | - J. Michael Gaziano
- Veterans Affairs Boston Healthcare System, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Sumitra Muralidhar
- Office of Research and Development, Department of Veterans Affairs, Washington, DC
| |
Collapse
|
18
|
Vasquez-Rios G, De Cos M, Campbell KN. Novel Therapies in APOL1-Mediated Kidney Disease: From Molecular Pathways to Therapeutic Options. Kidney Int Rep 2023; 8:2226-2234. [PMID: 38025220 PMCID: PMC10658239 DOI: 10.1016/j.ekir.2023.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 08/21/2023] [Indexed: 12/01/2023] Open
Abstract
Apolipoprotein L1 (APOL1) high-risk variants confer an increased risk for the development and progression of kidney disease among individuals of recent African ancestry. Over the past several years, significant progress has been made in understanding the pathogenesis of APOL1-mediated kidney diseases (AMKD), including genetic regulation, environmental interactions, immunomodulatory, proinflammatory and apoptotic signaling processes, as well as the complex role of APOL1 as an ion channel. Collectively, these findings have paved the way for novel therapeutic strategies to mitigate APOL1-mediated kidney injury. Precision medicine approaches are being developed to identify subgroups of AMKD patients who may benefit from these targeted interventions, fueling hope for improved clinical outcomes. This review summarizes key mechanistic insights in the pathogenesis of AMKD, emergent therapies, and discusses future challenges.
Collapse
Affiliation(s)
- George Vasquez-Rios
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marina De Cos
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirk N. Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
19
|
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. eLife 2023; 12:RP88538. [PMID: 37882666 PMCID: PMC10602586 DOI: 10.7554/elife.88538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023] Open
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 2242 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 2242 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]). Conclusions 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 (QF), R01HL163854 (QF), R35GM131770 (CMS), HL133786 (WQW), and Vanderbilt Faculty Research Scholar Fund (QF). 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
Affiliation(s)
- Lan Jiang
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Ge Liu
- Department of Biomedical Informatics, Vanderbilt University Medical CenterNashvilleUnited States
| | - Annette Oeser
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Andrea Ihegword
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Alyson L Dickson
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Laura L Daniel
- Division of Rheumatology, Department of Medicine, University of MiamiMiamiUnited States
| | - Adriana M Hung
- Tennessee Valley Healthcare System, Nashville CampusNashvilleUnited States
- Division of Nephrology & Hypertension, Vanderbilt University Medical CenterNashvilleUnited States
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| | - Cecilia P Chung
- Division of Rheumatology, Department of Medicine, University of MiamiMiamiUnited States
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical CenterNashvilleUnited States
| | - C Michael Stein
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
- Department of Pharmacology, Vanderbilt UniversityNashvilleUnited States
| | - Qiping Feng
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
- Vanderbilt Genetics Institute, Department of Medicine, Vanderbilt University Medical CenterNashvilleUnited States
| |
Collapse
|
20
|
Zhou X, Wang N, Liu W, Chen R, Yang G, Yu H. Identification of the potential association between SARS-CoV-2 infection and acute kidney injury based on the shared gene signatures and regulatory network. BMC Infect Dis 2023; 23:655. [PMID: 37789254 PMCID: PMC10548629 DOI: 10.1186/s12879-023-08638-6] [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: 04/18/2023] [Accepted: 09/24/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is identified as the cause of coronavirus disease 2019 (COVID-19) pandemic. Acute kidney injury (AKI), one of serious complications of COVID-19 infection, is the leading contributor to renal failure, associating with high mortality of the patients. This study aimed to identify the shared gene signatures and construct the gene regulatory network between COVID-19 and AKI, contributing to exploring the potential pathogenesis. METHODS Utilizing the machine learning approach, the candidate gene signatures were derived from the common differentially expressed genes (DEGs) obtained from COVID-19 and AKI. Subsequently, receiver operating characteristic (ROC), consensus clustering and functional enrichment analyses were performed. Finally, protein-protein interaction (PPI) network, transcription factor (TF)-gene interaction, gene-miRNA interaction, and TF-miRNA coregulatory network were systematically undertaken. RESULTS We successfully identified the shared 6 candidate gene signatures (RRM2, EGF, TMEM252, RARRES1, COL6A3, CUBN) between COVID-19 and AKI. ROC analysis showed that the model constructed by 6 gene signatures had a high predictive efficacy in COVID-19 (AUC = 0.965) and AKI (AUC = 0.962) cohorts, which had the potential to be the shared diagnostic biomarkers for COVID-19 and AKI. Additionally, the comprehensive gene regulatory networks, including PPI, TF-gene interaction, gene-miRNA interaction, and TF-miRNA coregulatory networks were displayed utilizing NetworkAnalyst platform. CONCLUSIONS This study successfully identified the shared gene signatures and constructed the comprehensive gene regulatory network between COVID-19 and AKI, which contributed to predicting patients' prognosis and providing new ideas for developing therapeutic targets for COVID-19 and AKI.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Nephrology, Haihe Hospital, Tianjin University, 890 Jingu Road, Jinnan District, Tianjin, 300350, China.
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, 300350, China.
- Haihe Clinical School, Tianjin Medical University, Tianjin, 300350, China.
- Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
| | - Ning Wang
- The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Wenjing Liu
- Department of Nephrology, Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Ruixue Chen
- Tianjin Haihe Hospital, Tianjin, 300350, China
| | - Guoyue Yang
- The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China.
| | - Hongzhi Yu
- Tianjin Institute of Respiratory Diseases, Tianjin, 300350, China.
- Department of Respiratory Medicine, Tianjin Haihe Hospital, 890 Jingu Road, Jinnan District, Tianjin, 300350, China.
| |
Collapse
|
21
|
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
|
22
|
Ramamoorthy R, Hussain H, Ravelo N, Sriramajayam K, Di Gregorio DM, Paulrasu K, Chen P, Young K, Masciarella AD, Jayakumar AR, Paidas MJ. Kidney Damage in Long COVID: Studies in Experimental Mice. BIOLOGY 2023; 12:1070. [PMID: 37626956 PMCID: PMC10452084 DOI: 10.3390/biology12081070] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023]
Abstract
Signs and symptoms involving multiple organ systems which persist for weeks or months to years after the initial SARS-CoV-2 infection (also known as PASC or long COVID) are common complications of individuals with COVID-19. We recently reported pathophysiological changes in various organs post-acute infection of mice with mouse hepatitis virus-1 (MHV-1, a coronavirus) (7 days) and after long-term post-infection (12 months). One of the organs severely affected in this animal model is the kidney, which correlated well with human studies showing kidney injury post-SARS-CoV-2 infection. Our long-term post-infection pathological observation in kidneys includes the development of edema and inflammation of the renal parenchyma, severe acute tubular necrosis, and infiltration of macrophages and lymphocytes, in addition to changes observed in both acute and long-term post-infection, which include tubular epithelial cell degenerative changes, peritubular vessel congestion, proximal and distal tubular necrosis, hemorrhage in the interstitial tissue, and vacuolation of renal tubules. These findings strongly suggest the possible development of renal fibrosis, in particular in the long-term post-infection. Accordingly, we investigated whether the signaling system that is known to initiate the above-mentioned changes in kidneys in other conditions is also activated in long-term post-MHV-1 infection. We found increased TGF-β1, FGF23, NGAL, IL-18, HIF1-α, TLR2, YKL-40, and B2M mRNA levels in long-term post-MHV-1 infection, but not EGFR, TNFR1, BCL3, and WFDC2. However, only neutrophil gelatinase-associated lipocalin (NGAL) increased in acute infection (7 days). Immunoblot studies showed an elevation in protein levels of HIF1-α, TLR-2, and EGFR in long-term post-MHV-1 infection, while KIM-1 and MMP-7 protein levels are increased in acute infection. Treatment with a synthetic peptide, SPIKENET (SPK), which inhibits spike protein binding, reduced NGAL mRNA in acute infection, and decreased TGF-β1, BCL3 mRNA, EGFR, HIF1-α, and TLR-2 protein levels long-term post-MHV-1 infection. These findings suggest that fibrotic events may initiate early in SARS-CoV-2 infection, leading to pronounced kidney fibrosis in long COVID. Targeting these factors therapeutically may prevent acute or long-COVID-associated kidney complications.
Collapse
Affiliation(s)
- Rajalakshmi Ramamoorthy
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.); (N.R.)
| | - Hussain Hussain
- Department of Internal Medicine and Infectious Disease, Larkin Community Hospital, Miami, FL 33143, USA;
| | - Natalia Ravelo
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.); (N.R.)
| | - Kannappan Sriramajayam
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Dibe M. Di Gregorio
- University of Miami College of Arts and Sciences, Coral Gables, FL 33146, USA;
| | - Kodisundaram Paulrasu
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (P.C.); (K.Y.)
| | - Karen Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (P.C.); (K.Y.)
| | | | - Arumugam R. Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.); (N.R.)
| | - Michael J. Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.); (N.R.)
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
23
|
Sen S, Khosla S, Awan O, Cohen S, Gollie JM. Endothelial dysfunction in autoimmune, pulmonary, and kidney systems, and exercise tolerance following SARS-CoV-2 infection. Front Med (Lausanne) 2023; 10:1197061. [PMID: 37575987 PMCID: PMC10413142 DOI: 10.3389/fmed.2023.1197061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Long COVID is characterized by persistent symptoms beyond 3-months of severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) infection that last for at least 2 months and cannot be explained by an alternative diagnosis. Autonomic, immunologic, endothelial, and hypercoagulation are implicated as possible mechanisms of long COVID symptoms. Despite recognition of the public health challenges posed by long COVID, the current understanding of the pathophysiological underpinnings is still evolving. In this narrative review, we explore the long-term effects of SARS-CoV-2 infection on T cell activation such as autoimmune disorders and endothelial cell dysfunction involving vascular impairments within pulmonary and renal architecture. We have described how endothelial dysfunction and vascular abnormalities may underscore findings of exercise intolerance by way of impaired peripheral oxygen extraction in individuals with long COVID.
Collapse
Affiliation(s)
- Sabyasachi Sen
- Division of Endocrinology, Department of Medicine, Veterans Affairs Medical Center, Washington, DC, United States
- Division of Endocrinology, Department of Medicine, George Washington University, Washington, DC, United States
| | - Shikha Khosla
- Division of Endocrinology, Department of Medicine, Veterans Affairs Medical Center, Washington, DC, United States
- Division of Endocrinology, Department of Medicine, George Washington University, Washington, DC, United States
| | - Omar Awan
- Division of Pulmonary Medicine, Department of Medicine, Veterans Affairs Medical Center, Washington, DC, United States
- Division of Pulmonary, Critical Care, and Sleep Disorders Medicine, The George Washington University, Washington, DC, United States
| | - Scott Cohen
- Division of Nephrology, Department of Medicine, Veterans Affairs Medical Center, Washington, DC, United States
| | - Jared M. Gollie
- Research and Development Service, Veterans Affairs Medical Center, Washington, DC, United States
- Department of Health, Human Function, and Rehabilitation Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
24
|
Grampp S, Krüger R, Lauer V, Uebel S, Knaup KX, Naas J, Höffken V, Weide T, Schiffer M, Naas S, Schödel J. Hypoxia hits APOL1 in the kidney. Kidney Int 2023; 104:53-60. [PMID: 37098381 DOI: 10.1016/j.kint.2023.03.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/02/2023] [Accepted: 03/24/2023] [Indexed: 04/27/2023]
Abstract
Individuals of African ancestry carrying two pathogenic variants of apolipoprotein 1 (APOL1) have a substantially increased risk for developing chronic kidney disease. The course of APOL1 nephropathy is extremely heterogeneous and shaped by systemic factors such as a response to interferon. However, additional environmental factors operating in this second-hit model have been less well defined. Here, we reveal that stabilization of hypoxia-inducible transcription factors (HIF) by hypoxia or HIF prolyl hydroxylase inhibitors activates transcription of APOL1 in podocytes and tubular cells. An active regulatory DNA-element upstream of APOL1 that interacted with HIF was identified. This enhancer was accessible preferentially in kidney cells. Importantly, upregulation of APOL1 by HIF was additive to the effects of interferon. Furthermore, HIF stimulated expression of APOL1 in tubular cells derived from the urine of an individual carrying a risk variant for kidney disease. Thus, hypoxic insults may serve as important modulators of APOL1 nephropathy.
Collapse
Affiliation(s)
- Steffen Grampp
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - René Krüger
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Victoria Lauer
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Uebel
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Karl X Knaup
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Naas
- Center for Integrative Bioinformatics Vienna, Max Perutz Labs, Vienna BioCenter, University of Vienna and Medical University of Vienna, Wien, Austria
| | - Verena Höffken
- Medical Clinic D, Institute of Molecular Nephrology, University Hospital of Münster, Münster, Germany
| | - Thomas Weide
- Medical Clinic D, Institute of Molecular Nephrology, University Hospital of Münster, Münster, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie Naas
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
25
|
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
|
26
|
Dashtban A, Mizani MA, Pasea L, Denaxas S, Corbett R, Mamza JB, Gao H, Morris T, Hemingway H, Banerjee A. Identifying subtypes of chronic kidney disease with machine learning: development, internal validation and prognostic validation using linked electronic health records in 350,067 individuals. EBioMedicine 2023; 89:104489. [PMID: 36857859 PMCID: PMC9989643 DOI: 10.1016/j.ebiom.2023.104489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Although chronic kidney disease (CKD) is associated with high multimorbidity, polypharmacy, morbidity and mortality, existing classification systems (mild to severe, usually based on estimated glomerular filtration rate, proteinuria or urine albumin-creatinine ratio) and risk prediction models largely ignore the complexity of CKD, its risk factors and its outcomes. Improved subtype definition could improve prediction of outcomes and inform effective interventions. METHODS We analysed individuals ≥18 years with incident and prevalent CKD (n = 350,067 and 195,422 respectively) from a population-based electronic health record resource (2006-2020; Clinical Practice Research Datalink, CPRD). We included factors (n = 264 with 2670 derived variables), e.g. demography, history, examination, blood laboratory values and medications. Using a published framework, we identified subtypes through seven unsupervised machine learning (ML) methods (K-means, Diana, HC, Fanny, PAM, Clara, Model-based) with 66 (of 2670) variables in each dataset. We evaluated subtypes for: (i) internal validity (within dataset, across methods); (ii) prognostic validity (predictive accuracy for 5-year all-cause mortality and admissions); and (iii) medications (new and existing by British National Formulary chapter). FINDINGS After identifying five clusters across seven approaches, we labelled CKD subtypes: 1. Early-onset, 2. Late-onset, 3. Cancer, 4. Metabolic, and 5. Cardiometabolic. Internal validity: We trained a high performing model (using XGBoost) that could predict disease subtypes with 95% accuracy for incident and prevalent CKD (Sensitivity: 0.81-0.98, F1 score:0.84-0.97). Prognostic validity: 5-year all-cause mortality, hospital admissions, and incidence of new chronic diseases differed across CKD subtypes. The 5-year risk of mortality and admissions in the overall incident CKD population were highest in cardiometabolic subtype: 43.3% (42.3-42.8%) and 29.5% (29.1-30.0%), respectively, and lowest in the early-onset subtype: 5.7% (5.5-5.9%) and 18.7% (18.4-19.1%). MEDICATIONS Across CKD subtypes, the distribution of prescription medication classes at baseline varied, with highest medication burden in cardiometabolic and metabolic subtypes, and higher burden in prevalent than incident CKD. INTERPRETATION In the largest CKD study using ML, to-date, we identified five distinct subtypes in individuals with incident and prevalent CKD. These subtypes have relevance to study of aetiology, therapeutics and risk prediction. FUNDING AstraZeneca UK Ltd, Health Data Research UK.
Collapse
Affiliation(s)
- Ashkan Dashtban
- Institute of Health Informatics, University College London, London, UK
| | - Mehrdad A Mizani
- Institute of Health Informatics, University College London, London, UK; British Heart Foundation Data Science Centre, Health Data Research UK, London, UK
| | - Laura Pasea
- Institute of Health Informatics, University College London, London, UK
| | - Spiros Denaxas
- Institute of Health Informatics, University College London, London, UK
| | | | - Jil B Mamza
- Medical and Scientific Affairs, BioPharmaceuticals Medical, AstraZeneca, London, UK
| | - He Gao
- Medical and Scientific Affairs, BioPharmaceuticals Medical, AstraZeneca, London, UK
| | - Tamsin Morris
- Medical and Scientific Affairs, BioPharmaceuticals Medical, AstraZeneca, London, UK
| | - Harry Hemingway
- Institute of Health Informatics, University College London, London, UK; Health Data Research UK, University College London, London, UK
| | - Amitava Banerjee
- Institute of Health Informatics, University College London, London, UK; Barts Health NHS Trust, London, UK; University College London Hospitals NHS Trust, London, UK.
| |
Collapse
|
27
|
Wan ES, Verma A, Minnier J, Luoh SW, Iyengar SK. Reply to: Sookaromdee and Wiwanitkit. Am J Respir Crit Care Med 2023; 207:113-114. [PMID: 36041210 PMCID: PMC9952861 DOI: 10.1164/rccm.202208-1611le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Emily S. Wan
- VA Boston Healthcare SystemBoston, Massachusetts,Brigham and Women’s HospitalBoston, Massachusetts
| | - Anurag Verma
- Corporal Michael J Crescenz VA Medical CenterPhiladelphia, Pennsylvania
| | - Jessica Minnier
- VA Portland Health Care SystemPortland, Oregon,Louis Stokes Cleveland VA Medical CenterCleveland, Ohio
| | - Shiuh-Wen Luoh
- VA Portland Health Care SystemPortland, Oregon,Oregon Health and Science UniversityPortland, Oregon,Corresponding author (e-mail: )
| | | |
Collapse
|
28
|
Harley JB, Pyarajan S, Partan ES, Epstein L, Wertheim JA, Diwan A, Woods CW, Davey V, Blair S, Clark DH, Kaufman KM, Khan S, Chepelev I, Devine A, Cameron P, McCann MF, Ammons MCB, Bolz DD, Battles JK, Curtis JL, Holodniy M, Marconi VC, Searles CD, Beenhouwer DO, Brown ST, Moorman JP, Yao ZQ, Rodriguez-Barradas MC, Mohapatra S, Molina De Rodriguez OY, Padiernos EB, McIndoo ER, Price E, Burgoyne HM, Robey I, Schwenke DC, Shive CL, Przygodzki RM, Ramoni RB, Krull HK, Bonomo RA. The US Department of Veterans Affairs Science and Health Initiative to Combat Infectious and Emerging Life-Threatening Diseases (VA SHIELD): A Biorepository Addressing National Health Threats. Open Forum Infect Dis 2022; 9:ofac641. [PMID: 36601554 PMCID: PMC9801224 DOI: 10.1093/ofid/ofac641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Indexed: 12/15/2022] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has demonstrated the need to share data and biospecimens broadly to optimize clinical outcomes for US military Veterans. Methods In response, the Veterans Health Administration established VA SHIELD (Science and Health Initiative to Combat Infectious and Emerging Life-threatening Diseases), a comprehensive biorepository of specimens and clinical data from affected Veterans to advance research and public health surveillance and to improve diagnostic and therapeutic capabilities. Results VA SHIELD now comprises 12 sites collecting de-identified biospecimens from US Veterans affected by SARS-CoV-2. In addition, 2 biorepository sites, a data processing center, and a coordinating center have been established under the direction of the Veterans Affairs Office of Research and Development. Phase 1 of VA SHIELD comprises 34 157 samples. Of these, 83.8% had positive tests for SARS-CoV-2, with the remainder serving as contemporaneous controls. The samples include nasopharyngeal swabs (57.9%), plasma (27.9%), and sera (12.5%). The associated clinical and demographic information available permits the evaluation of biological data in the context of patient demographics, clinical experience and management, vaccinations, and comorbidities. Conclusions VA SHIELD is representative of US national diversity with a significant potential to impact national healthcare. VA SHIELD will support future projects designed to better understand SARS-CoV-2 and other emergent healthcare crises. To the extent possible, VA SHIELD will facilitate the discovery of diagnostics and therapeutics intended to diminish COVID-19 morbidity and mortality and to reduce the impact of new emerging threats to the health of US Veterans and populations worldwide.
Collapse
Affiliation(s)
- John B Harley
- Correspondence: John B. Harley, Cincinnati VA Medical Center, 3200 Vine St., John B. Harley (151), Cincinnati, OH 45220 ()
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
| | - Elizabeth S Partan
- Center for Data and Computational Sciences, Veterans Affairs Boston Healthcare System, Boston, Massachusetts, USA
| | - Lauren Epstein
- Infectious Diseases, US Department of Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - Jason A Wertheim
- Research & Development, Southern Arizona Veterans Affairs Healthcare System, US Department of Veterans Affairs, Tucson, Arizona, USA
| | - Abhinav Diwan
- Cardiology, Veterans Affairs Saint Louis Healthcare System, US Department of Veterans Affairs,Saint Louis, Missouri, USA
| | - Christopher W Woods
- Medicine, US Department of Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Victoria Davey
- Office of Research and Development, US Department of Veterans Affairs, Washington, District of Columbia, USA
| | - Sharlene Blair
- Research Services, US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Dennis H Clark
- Research Services, US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Kenneth M Kaufman
- Research Services, US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Shagufta Khan
- Research Services, US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Iouri Chepelev
- Research Services, US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Alexander Devine
- Prometheus Federal Services, Titan Alpha, Washington, District of Columbia, USA
| | - Perry Cameron
- Customer Value Partners, Titan Alpha, Washington, District of Columbia, USA
| | - Monica F McCann
- Office of Research and Development, Chesapeake Medical Communications, Contractor for the US Department of Veterans Affairs, Washington, District of Columbia, USA
| | - Mary Cloud B Ammons
- Research, US Department of Veterans Affairs Medical Center, Boise, Idaho, USA,Idaho Veterans Research and Education Foundation, Boise, Idaho, USA
| | - Devin D Bolz
- Research, US Department of Veterans Affairs Medical Center, Boise, Idaho, USA
| | - Jane K Battles
- Office of Research and Development, US Department of Veterans Affairs, Washington, District of Columbia, USA
| | - Jeffrey L Curtis
- Medicine Service, Veteran Affairs Ann Arbor Healthcare System, US Department of Veterans Affairs, Ann Arbor, Michigan, USA
| | - Mark Holodniy
- Public Health Surveillance, Veterans Affairs Palo Alto Healthcare System, US Department of Veterans Affairs, Palo Alto, California, USA
| | - Vincent C Marconi
- Infectious Diseases, US Department of Veterans Affairs Medical Center, Atlanta, Georgia, USA,Division of Infectious Diseases, Emory School of Medicine and Rollins School of Public Health, Atlanta, Georgia, USA
| | - Charles D Searles
- Infectious Diseases, US Department of Veterans Affairs Medical Center, Atlanta, Georgia, USA
| | - David O Beenhouwer
- Medicine, Veterans Affairs Greater Los Angeles Healthcare System, US Department of Veterans Affairs, Los Angeles, California, USA
| | - Sheldon T Brown
- Infectious Diseases, James J. Peters Veterans Affairs Medical Center, US Department of Veterans Affairs, Bronx, New York, USA
| | - Jonathan P Moorman
- Infectious Diseases, James H. Quillen Veterans Affairs Medical Center, US Department of Veterans Affairs, Mountain Home, Tennessee, USA,Center of Excellence in Inflammation, Infectious Diseases, and Immunity, East Tennessee State University, Johnson City, Tennessee, USA
| | - Zhi Q Yao
- Infectious Diseases, James H. Quillen Veterans Affairs Medical Center, US Department of Veterans Affairs, Mountain Home, Tennessee, USA,Center of Excellence in Inflammation, Infectious Diseases, and Immunity, East Tennessee State University, Johnson City, Tennessee, USA
| | - Maria C Rodriguez-Barradas
- Infectious Diseases Section, Michael E. DeBakey Veterans Affairs Medical Center, US Department of Veterans Affairs, Houston, Texas, USA,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Shyam Mohapatra
- Medicine, James A. Haley Veterans Hospital, US Department of Veterans Affairs, Tampa, Florida, USA
| | - Osmara Y Molina De Rodriguez
- Research & Development, Southern Arizona Veterans Affairs Healthcare System, US Department of Veterans Affairs, Tucson, Arizona, USA
| | - Emerson B Padiernos
- Research, US Department of Veterans Affairs Medical Center, Boise, Idaho, USA
| | - Eric R McIndoo
- Research, US Department of Veterans Affairs Medical Center, Boise, Idaho, USA,Idaho Veterans Research and Education Foundation, Boise, Idaho, USA
| | - Emily Price
- Research, US Department of Veterans Affairs Medical Center, Boise, Idaho, USA,Idaho Veterans Research and Education Foundation, Boise, Idaho, USA
| | - Hailey M Burgoyne
- Research, US Department of Veterans Affairs Medical Center, Boise, Idaho, USA,Idaho Veterans Research and Education Foundation, Boise, Idaho, USA
| | - Ian Robey
- Research & Development, Southern Arizona Veterans Affairs Healthcare System, US Department of Veterans Affairs, Tucson, Arizona, USA
| | - Dawn C Schwenke
- Research & Development, Southern Arizona Veterans Affairs Healthcare System, US Department of Veterans Affairs, Tucson, Arizona, USA
| | - Carey L Shive
- Medicine, Veterans Affairs Northeast Ohio Healthcare System, US Department of Veterans Affairs, Cleveland, Ohio, USA
| | - Ronald M Przygodzki
- Office of Research and Development, US Department of Veterans Affairs, Washington, District of Columbia, USA
| | - Rachel B Ramoni
- Office of Research and Development, US Department of Veterans Affairs, Washington, District of Columbia, USA
| | | | | |
Collapse
|
29
|
Geetha D, Kronbichler A, Rutter M, Bajpai D, Menez S, Weissenbacher A, Anand S, Lin E, Carlson N, Sozio S, Fowler K, Bignall R, Ducharlet K, Tannor EK, Wijewickrama E, Hafidz MIA, Tesar V, Hoover R, Crews D, Varnell C, Danziger-Isakov L, Jha V, Mohan S, Parikh C, Luyckx V. Impact of the COVID-19 pandemic on the kidney community: lessons learned and future directions. Nat Rev Nephrol 2022; 18:724-737. [PMID: 36002770 PMCID: PMC9400561 DOI: 10.1038/s41581-022-00618-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2022] [Indexed: 12/15/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has disproportionately affected patients with kidney disease, causing significant challenges in disease management, kidney research and trainee education. For patients, increased infection risk and disease severity, often complicated by acute kidney injury, have contributed to high mortality. Clinicians were faced with high clinical demands, resource shortages and novel ethical dilemmas in providing patient care. In this review, we address the impact of COVID-19 on the entire spectrum of kidney care, including acute kidney injury, chronic kidney disease, dialysis and transplantation, trainee education, disparities in health care, changes in health care policies, moral distress and the patient perspective. Based on current evidence, we provide a framework for the management and support of patients with kidney disease, infection mitigation strategies, resource allocation and support systems for the nephrology workforce.
Collapse
Affiliation(s)
- Duvuru Geetha
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | - Megan Rutter
- Department of Lifespan and Population Health, University of Nottingham, Nottingham, UK
| | - Divya Bajpai
- Department of Nephrology, Seth Gordhandas Sunderdas Medical College (GSMC) and the King Edward Memorial (KEM) Hospital, Mumbai, India
| | - Steven Menez
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Annemarie Weissenbacher
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Shuchi Anand
- Department of Medicine, Division of Nephrology, Stanford University School of Medicine, Standford, California, USA
| | - Eugene Lin
- Department of Internal Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Leonard D. Schaeffer Center for Health Policy & Economics, University of Southern California, Los Angeles, California, USA
| | - Nicholas Carlson
- Department of Nephrology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Research, The Danish Heart Foundation, Copenhagen, Denmark
| | - Stephen Sozio
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin Fowler
- Principal, Voice of the Patient Inc, St. Louis, Missouri, USA
| | - Ray Bignall
- Division of Nephrology and Hypertension, Nationwide Children's Hospital and Department of Paediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Kathryn Ducharlet
- Department of Renal Medicine, Eastern Health Clinical School, Monash University, Melbourne, Australia
- Department of Nephrology and Palliative Care, St Vincent's Hospital Melbourne, Australia and Department of Medicine, University of Melbourne, Parkville, Australia
| | - Elliot K Tannor
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Renal Unit, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Eranga Wijewickrama
- Consultant Nephrologist and Professor in the Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
- University Medical Unit, National Hospital of Sri Lanka and National Institute of Nephrology, Dialysis & Transplantation, Colombo, Sri Lanka
| | | | - Vladimir Tesar
- Department of Nephrology, Charles University, Prague, Czech Republic
| | - Robert Hoover
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Deidra Crews
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles Varnell
- Division of Nephrology & Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- James M. Anderson Center for Health Systems Excellence, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Paediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio, USA
| | - Lara Danziger-Isakov
- Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio, USA
| | - Vivekanand Jha
- George Institute for Global Health, New Delhi, India
- School of Public Health, Imperial College, London, UK
- Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, India
| | - Sumit Mohan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons and the Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, USA
| | - Chirag Parikh
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Valerie Luyckx
- Associate Scientist, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
- Honorary Associate Professor, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
- Nephrologist, University Childrens Hospital, Zurich, Switzerland
| |
Collapse
|
30
|
Hassan MO, Balogun RA. The Effects of Race on Acute Kidney Injury. J Clin Med 2022; 11:5822. [PMID: 36233687 PMCID: PMC9573379 DOI: 10.3390/jcm11195822] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
Racial disparities in incidence and outcomes of acute kidney injury (AKI) are pervasive and are driven in part by social inequities and other factors. It is well-documented that Black patients face higher risk of AKI and seemingly have a survival advantage compared to White counterparts. Various explanations have been advanced and suggested to account for this, including differences in susceptibility to kidney injury, severity of illness, and socioeconomic factors. In this review, we try to understand and further explore the link between race and AKI using the incidence, diagnosis, and management of AKI to illustrate how race is directly related to AKI outcomes, with a focus on Black and White individuals with AKI. In particular, we explore the effect of race-adjusted estimated glomerular filtration rate (eGFR) equation on AKI prediction and discuss racial disparities in the management of AKI and how this might contribute to racial differences in AKI-related mortality among Blacks with AKI. We also identify some opportunities for future research and advocacy.
Collapse
Affiliation(s)
- Muzamil Olamide Hassan
- Department of Medicine, Obafemi Awolowo University, Ile-Ife 220005, Nigeria
- Division of Nephrology, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Rasheed Abiodun Balogun
- Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
31
|
Yang L, Li J, Wei W, Yi C, Pu Y, Zhang L, Cui T, Ma L, Zhang J, Koyner J, Zhao Y, Fu P. Kidney health in the COVID-19 pandemic: An umbrella review of meta-analyses and systematic reviews. Front Public Health 2022; 10:963667. [PMID: 36172213 PMCID: PMC9511113 DOI: 10.3389/fpubh.2022.963667] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/16/2022] [Indexed: 01/24/2023] Open
Abstract
Background This umbrella review aims to consolidate evidence from systematic reviews and meta-analyses investigating the impact of the coronavirus disease-2019 (COVID-19) on kidney health, and the associations between kidney diseases and clinical outcomes in COVID-19 patients. Methods Five databases, namely, EMBASE, PubMed, Web of Science, the Cochrane Database of Systematic Reviews and Ovid Medline, were searched for meta-analyses and systematic reviews from January 1, 2020 to June 2, 2022. Two reviewers independently selected reviews, identified reviews for inclusion and extracted data. Disagreements were resolved by group discussions. Two reviewers independently assessed the methodological quality of all included reviews using ROBIS tool. A narrative synthesis was conducted. The characteristics and major findings of the included reviews are presented using tables and forest plots. The included meta-analyses were updated when necessary. The review protocol was prospectively registered in PROSPERO (CRD42021266300). Results A total of 103 reviews were identified. Using ROBIS, 30 reviews were rated as low risk of bias. Data from these 30 reviews were included in the narrative synthesis. Ten meta-analyses were updated by incorporating 119 newly available cohort studies. Hospitalized COVID-19 patients had a notable acute kidney injury (AKI) incidence of 27.17%. AKI was significantly associated with mortality (pooled OR: 5.24) and severe conditions in COVID-19 patients (OR: 14.94). The pooled prevalence of CKD in COVID-19 patients was 5.7%. Pre-existing CKD was associated with a higher risk of death (pooled OR: 2.21) and disease severity (pooled OR: 1.87). Kidney transplant recipients were susceptible to SARS-CoV-2 infection (incidence: 23 per 10,000 person-weeks) with a pooled mortality of 18%. Conclusion Kidney disease such as CKD or recipients of kidney transplants were at increased risk of contracting COVID-19. Persons with COVID-19 also had a notable AKI incidence. AKI, the need for RRT, pre-existing CKD and a history of kidney transplantation are associated with adverse outcomes in COVID-19. Systematic review registration www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021266300, identifier: CRD42021266300.
Collapse
Affiliation(s)
- Letian Yang
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Jian Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Wei Wei
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Cheng Yi
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yajun Pu
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Ling Zhang
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Tianlei Cui
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Liang Ma
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Juqian Zhang
- Liverpool Centre for Cardiovascular Science, Liverpool Heart and Chest Hospital, University of Liverpool, Liverpool, United Kingdom
| | - Jay Koyner
- Division of Nephrology, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Yuliang Zhao
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Ping Fu
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Abstract
The acute coronavirus disease-2019 (COVID-19) pandemic has had a significant impact on the incidence and prevalence of acute kidney injury and chronic kidney disease globally and in low-income settings. Chronic kidney disease increases the risk of developing COVID-19 and COVID-19 causes acute kidney injury directly or indirectly and is associated with high mortality in severe cases. Outcomes of COVID-19-associated kidney disease were not equitable globally owing to a lack of health infrastructure, challenges in diagnostic testing, and management of COVID-19 in low-income settings. COVID-19 also significantly impacted kidney transplant rates and mortality among kidney transplant recipients. Vaccine availability and uptake remains a significant challenge in low- and lower-middle-income countries compared with high-income countries. In this review, we explore the inequities in low- and lower-middle-income countries and highlight the progress made in the prevention, diagnosis, and management of patients with COVID-19 and kidney disease. We recommend further studies into the challenges, lessons learned, and progress made in the diagnosis, management, and treatment of patients with COVID-19-related kidney diseases and suggest ways to improve the care and management of patients with COVID-19 and kidney disease.
Collapse
|
33
|
Sukumar S, Orkaby AR, Schwartz JB, Marcum Z, Januzzi JL, Vaduganathan M, Warraich HJ. Polypharmacy in Older Heart Failure Patients: a Multidisciplinary Approach. Curr Heart Fail Rep 2022; 19:290-302. [PMID: 35723783 DOI: 10.1007/s11897-022-00559-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/02/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW We provide a review of considerations when applying principles of optimal pharmacotherapy to older adults with heart failure (HF), an analysis on the pivotal clinical trials focusing on applicability to older adults, and multi-disciplinary strategies to optimize the health of HF patients with polypharmacy. RECENT FINDINGS Polypharmacy is very common among patients with HF, due to medications for both HF and non-HF comorbidities. Definitions of polypharmacy were not developed specifically for older adults with HF and may need to be modified in order to meaningfully describe medication burden and promote appropriate medical therapy. This is because clinical practice guidelines for multi-drug HF regimens have unique considerations, given that they improve outcomes and symptoms of HF. Adults older than 65 years are well represented in contemporary clinical trials for HF with preserved ejection fraction (HFpEF) and guideline directed medical therapy (GDMT) for HF with reduced ejection fraction (HFrEF). While these trials did not have significant heterogeneity in safety or efficacy across a broad age spectrum, some may have limited representation of adults ≥ 80 years old, the sickest older adults, or those with decreased functional status. There is also a lack of data on the safety and efficacy of deprescribing HF medications, and deprescription in otherwise stable patients may lead to clinical destabilization or disease progression. There is therefore innate tension between the well-studied benefits of optimized HF therapy for older adults that must be weighed against the risks of polypharmacy and many unknowns that still exist. Given the strong evidence that optimized HF therapies confer symptomatic and mortality benefits for older adults, it is clear that polypharmacy in this context can be appropriate. A shift in paradigm is therefore needed when evaluating polypharmacy in patients with HF. Instead of assuming all polypharmacy is "good" or "bad," we propose a concerted move, using a multidisciplinary approach, to focus on the "appropriateness" of specific medications, in order to optimize HF medical therapy. Clinicians of all specialties caring for complex older adults with HF must consider goals of care, functional status, and new evidence-based therapies, in order to optimize this polypharmacy for older adults.
Collapse
Affiliation(s)
- Smrithi Sukumar
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ariela R Orkaby
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA.,New England Geriatric Research, Education, and Clinical Center (GRECC), VA Boston Healthcare System, Boston, MA, USA.,Division of Aging, Brigham & Women's Hospital, Boston, MA, USA
| | | | - Zachary Marcum
- UW School of Pharmacy, University of Washington, Seattle, WA, USA
| | - James L Januzzi
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Muthiah Vaduganathan
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Haider J Warraich
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA.,Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA.,, Boston, MA, USA
| |
Collapse
|
34
|
He W, Liu X, Hu B, Li D, Chen L, Li Y, Tu Y, Xiong S, Wang G, Deng J, Fu B. Mechanisms of SARS-CoV-2 Infection-Induced Kidney Injury: A Literature Review. Front Cell Infect Microbiol 2022; 12:838213. [PMID: 35774397 PMCID: PMC9237415 DOI: 10.3389/fcimb.2022.838213] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/10/2022] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory coronavirus 2 (SARS-CoV-2) has become a life-threatening pandemic. Clinical evidence suggests that kidney involvement is common and might lead to mild proteinuria and even advanced acute kidney injury (AKI). Moreover, AKI caused by coronavirus disease 2019 (COVID-19) has been reported in several countries and regions, resulting in high patient mortality. COVID-19-induced kidney injury is affected by several factors including direct kidney injury mediated by the combination of virus and angiotensin-converting enzyme 2, immune response dysregulation, cytokine storm driven by SARS-CoV-2 infection, organ interactions, hypercoagulable state, and endothelial dysfunction. In this review, we summarized the mechanism of AKI caused by SARS-CoV-2 infection through literature search and analysis.
Collapse
Affiliation(s)
- Weihang He
- Reproductive Medicine Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqiang Liu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Bing Hu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dongshui Li
- Reproductive Medicine Center, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Luyao Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yu Li
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yechao Tu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Situ Xiong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Jun Deng
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Fu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| |
Collapse
|
35
|
Wu M, Chen Y, Chiu I, Wu M. Genetic Insight into Primary Glomerulonephritis. Nephrology (Carlton) 2022; 27:649-657. [DOI: 10.1111/nep.14074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/18/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Mei‐Yi Wu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health National Taiwan University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
| | - Ying‐Chun Chen
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
| | - I‐Jen Chiu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
| | - Mai‐Szu Wu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine Taipei Medical University Taipei Taiwan
| |
Collapse
|
36
|
Biancolella M, Colona VL, Mehrian-Shai R, Watt JL, Luzzatto L, Novelli G, Reichardt JKV. COVID-19 2022 update: transition of the pandemic to the endemic phase. Hum Genomics 2022; 16:19. [PMID: 35650595 PMCID: PMC9156835 DOI: 10.1186/s40246-022-00392-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19, which is caused by the SARS-CoV-2, has ravaged the world for the past 2 years. Here, we review the current state of research into the disease with focus on its history, human genetics and genomics and the transition from the pandemic to the endemic phase. We are particularly concerned by the lack of solid information from the initial phases of the pandemic that highlighted the necessity for better preparation to face similar future threats. On the other hand, we are gratified by the progress into human genetic susceptibility investigations and we believe now is the time to explore the transition from the pandemic to the endemic phase. The latter will require worldwide vigilance and cooperation, especially in emerging countries. In the transition to the endemic phase, vaccination rates have lagged and developed countries should assist, as warranted, in bolstering vaccination rates worldwide. We also discuss the current status of vaccines and the outlook for COVID-19.
Collapse
Affiliation(s)
| | - Vito Luigi Colona
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133, Rome, Italy
| | - Ruty Mehrian-Shai
- Sheba Medical Center, Pediatric Hemato-Oncology, Edmond and Lilly Safra Children's Hospital, Tel Hashomer 2 Sheba Road, 52621, Ramat Gan, Israel
| | - Jessica Lee Watt
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Smithfield, QLD, 4878, Australia
| | - Lucio Luzzatto
- Department of Haematology and Blood Transfusion, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania.,University of Florence, Florence, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133, Rome, Italy. .,IRCCS Neuromed, Pozzilli, Isernia, Italy. .,Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV, USA. .,Department of Biomedicine and Prevention, School of Medicine and Surgery, Via Montpellier 1, 00133, Rome, Italy.
| | - Juergen K V Reichardt
- Australian Institute of Tropical Health and Medicine, James Cook University, Smithfield, QLD, 4878, Australia
| |
Collapse
|
37
|
Abstract
Kidney involvement is common in coronavirus disease-2019 (COVID-19), and our understanding of the effects of COVID-19 on short- and long-term kidney outcomes has evolved over the course of the pandemic. Initial key questions centered on the spectrum and degree of acute kidney injury (AKI) in patients hospitalized with severe COVID-19. Investigators worldwide have explored the association between COVID-19-associated AKI and short-term outcomes, including inpatient mortality and disease severity. Even as treatments evolved, vaccinations were developed, and newer viral variants arose, subsets of patients were identified as at continued high risk for major adverse kidney outcomes. In this review, we explore key topics of continued relevance including the following: (1) a comparison of COVID-19-associated AKI with AKI developing in other clinical settings; (2) the ongoing controversy over kidney tropism in the setting of COVID-19 and the potential for competitive binding of the severe acute respiratory syndrome coronavirus 2 virus with angiotensin converting enzyme-2 to prevent viral cell entry; and (3) the identification of high-risk patients for adverse outcomes to inform long-term outpatient management. Patients at particularly high risk for adverse kidney outcomes include those with APOL1 high-risk genotype status. Biomarkers of injury, inflammation, tubular health, and repair measured in both the blood and urine may hold prognostic significance.
Collapse
Affiliation(s)
| | - Chirag R. Parikh
- Address reprint requests to Chirag R. Parikh, MD, PhD, Division of Nephrology, Johns Hopkins University School of Medicine, 1830 E Monument St, Suite 416, Baltimore, MD 21287
| |
Collapse
|
38
|
Reiser J, Spear R, Luo S. SARS-CoV-2 pirates the kidneys: A scar(y) story. Cell Metab 2022; 34:352-354. [PMID: 35235772 PMCID: PMC8886684 DOI: 10.1016/j.cmet.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
SARS-CoV-2 can cause diverse severe and lasting damage to the kidneys. In the latest issue of Cell Stem Cell, Jansen et al. utilized data gleaned from human kidney autopsies and human induced pluripotent stem cell-derived kidney organoids to investigate the direct effects of SARS-CoV-2 infection on kidney cells. They found that such infections resulted in renal scarring (notably, tubulointerstitial fibrosis).
Collapse
Affiliation(s)
- Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, IL 60612, USA.
| | - Ryan Spear
- Department of Internal Medicine, Rush University, Chicago, IL 60612, USA
| | - Shengyuan Luo
- Department of Internal Medicine, Rush University, Chicago, IL 60612, USA
| |
Collapse
|