1
|
Jonusaite S, Himmerkus N. Paracellular barriers: Advances in assessing their contribution to renal epithelial function. Comp Biochem Physiol A Mol Integr Physiol 2024; 298:111741. [PMID: 39276851 DOI: 10.1016/j.cbpa.2024.111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Regulation of salt and water balance occupies a dominant role in the physiology of many animals and often relies on the function of the renal system. In the mammalian kidney, epithelial ion and water transport requires high degree of coordination between the transcellular and paracellular pathways, the latter being defined by the intercellular tight junctions (TJs). TJs seal the paracellular pathway in a highly specialized manner, either by forming a barrier against the passage of solutes and/or water or by allowing the passage of ions and/or water through them. This functional TJ plasticity is now known to be provided by the members of the claudin family of tetraspan proteins. Unlike mammalian nephron, the renal structures of insects, the Malpighian tubules, lack TJs and instead have smooth septate junctions (sSJs) as paracellular barrier forming junctions. Many questions regarding the molecular and functional properties of sSJs remain open but research on model species have begun to inform our understanding. The goal of this commentary is to highlight key concepts and most recent findings that have emerged from the molecular and functional dissection of paracellular barriers in the mammalian and insect renal epithelia.
Collapse
Affiliation(s)
- Sima Jonusaite
- Institute of Physiology, Christian-Albrechts-University of Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany.
| | - Nina Himmerkus
- Institute of Physiology, Christian-Albrechts-University of Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany
| |
Collapse
|
2
|
Vorperian SK, DeFelice BC, Buonomo JA, Chinchinian HJ, Gray IJ, Yan J, Mach KE, La V, Lee TJ, Liao JC, Lafayette R, Loeb GB, Bertozzi CR, Quake SR. Deconvolution of Human Urine across the Transcriptome and Metabolome. Clin Chem 2024; 70:1344-1354. [PMID: 39383112 DOI: 10.1093/clinchem/hvae137] [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: 04/18/2024] [Accepted: 07/10/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Early detection of the cell type changes underlying several genitourinary tract diseases largely remains an unmet clinical need, where existing assays, if available, lack the cellular resolution afforded by an invasive biopsy. While messenger RNA in urine could reflect the dynamic signal that facilitates early detection, current measurements primarily detect single genes and thus do not reflect the entire transcriptome and the underlying contributions of cell type-specific RNA. METHODS We isolated and sequenced the cell-free RNA (cfRNA) and sediment RNA from human urine samples (n = 6 healthy controls and n = 12 kidney stone patients) and measured the urine metabolome. We analyzed the resulting urine transcriptomes by deconvolving the noninvasively measurable cell type contributions and comparing to plasma cfRNA and the measured urine metabolome. RESULTS Urine transcriptome cell type deconvolution primarily yielded relative fractional contributions from genitourinary tract cell types in addition to cell types from high-turnover solid tissues beyond the genitourinary tract. Comparison to plasma cfRNA yielded enrichment of metabolic pathways and a distinct cell type spectrum. Integration of urine transcriptomic and metabolomic measurements yielded enrichment for metabolic pathways involved in amino acid metabolism and overlapped with metabolic subsystems associated with proximal tubule function. CONCLUSIONS Noninvasive whole transcriptome measurements of human urine cfRNA and sediment RNA reflects signal from hard-to-biopsy tissues exhibiting low representation in blood plasma cfRNA liquid biopsy at cell type resolution and are enriched in signal from metabolic pathways measurable in the urine metabolome.
Collapse
Affiliation(s)
- Sevahn K Vorperian
- Department of Chemical Engineering, Stanford University, Stanford, CA, United States
- Sarafan ChEM-H, Stanford University, Stanford, CA, United States
| | | | - Joseph A Buonomo
- Sarafan ChEM-H, Stanford University, Stanford, CA, United States
- Department of Chemistry, Stanford University, Stanford, CA, United States
| | - Hagop J Chinchinian
- Department of Electrical Engineering, Stanford University, Stanford, CA, United States
| | - Ira J Gray
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Jia Yan
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Kathleen E Mach
- Department of Urology, Stanford University, Stanford, CA, United States
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Vinh La
- Department of Urology, Stanford University, Stanford, CA, United States
| | - Timothy J Lee
- Department of Urology, Stanford University, Stanford, CA, United States
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Joseph C Liao
- Department of Urology, Stanford University, Stanford, CA, United States
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, United States
| | - Richard Lafayette
- Division of Nephrology, Stanford School of Medicine, Stanford, CA, United States
| | - Gabriel B Loeb
- Department of Medicine and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Carolyn R Bertozzi
- Sarafan ChEM-H, Stanford University, Stanford, CA, United States
- Department of Chemistry, Stanford University, Stanford, CA, United States
- Howard Hughes Medical Institute, Stanford, CA, United States
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, United States
- Department of Applied Physics, Stanford University, Stanford, CA, United States
- Chan Zuckerberg Initiative, Redwood City, CA, United States
| |
Collapse
|
3
|
Rousing AQ, Jeppesen M, Jensen-Fangel S, Leipziger J, Sorensen MV, Berg P. The challenged urine bicarbonate excretion test in cystic fibrosis: A comprehensive analysis of urine acid/base parameters. Acta Physiol (Oxf) 2024; 240:e14233. [PMID: 39308271 DOI: 10.1111/apha.14233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024]
Abstract
AIM Renal excretion of excess HCO3 - depends on renal cystic fibrosis transmembrane conductance regulator (CFTR) and is impaired in people with cystic fibrosis (pwCF). Urine HCO3 - excretion following oral NaHCO3-loading may be a simple in vivo biomarker of CFTR function. In this study, we investigated changes in urine acid/base parameters following oral NaHCO3-loading to comprehensively assess the physiological response to the test and evaluate HCO3 - as the primary test result. METHODS Urine acid/base parameters (titratable acid (TA), NH4 +, net acid excretion (NAE) and pH) were measured in bio-banked urine samples from controls (n = 10) and pwCF (n = 50) who completed the challenged urine HCO3 - test. The association between urine acid/base excretion parameters and clinical CF disease characteristics and CFTR modulator therapy-induced changes were assessed. RESULTS Before treatment, challenged urine acid/base excretion associated with important CF disease characteristics. TA excretion and NAE were lower in pwCF with residual function mutations, 7.9 and 16.6 mmol/3 h, respectively, and lower TA excretion and NAE associated with pancreatic sufficiency. A lower excretion of TA, NH4 +, and NAE associated with a higher percentage of predicted FEV1 (1.3%, 2.5% and 0.8% per mmol/3 h higher, respectively). Modulator treatment decreased TA excretion and NAE (-2.9 and -5.3 mmol/3 h, respectively). CONCLUSION Following acute NaHCO3-loading, increased base excretion is mirrored by decreased acid excretion. Urine HCO3 - excretion sufficiently represents the additional urine acid/base parameters as test result. The observed changes in acid excretion support CFTR modulator-induced increase of CFTR-dependent type B intercalated cell HCO3 - secretion and the use of the challenged urine HCO3 - test as a possible CFTR-biomarker.
Collapse
Affiliation(s)
| | - Majbritt Jeppesen
- Department of Infectious Diseases, Cystic Fibrosis Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Søren Jensen-Fangel
- Department of Infectious Diseases, Cystic Fibrosis Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Leipziger
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mads V Sorensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Peder Berg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
4
|
Kulow VA, Roegner K, Labes R, Kasim M, Mathia S, Czopek CS, Berndt N, Becker PN, Ter-Avetisyan G, Luft FC, Enghard P, Hinze C, Klocke J, Eckardt KU, Schmidt-Ott KM, Persson PB, Rosenberger C, Fähling M. Beyond hemoglobin: Critical role of 2,3-bisphosphoglycerate mutase in kidney function and injury. Acta Physiol (Oxf) 2024:e14242. [PMID: 39422260 DOI: 10.1111/apha.14242] [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: 08/14/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
AIM 2,3-bisphosphoglycerate mutase (BPGM) is traditionally recognized for its role in modulating oxygen affinity to hemoglobin in erythrocytes. Recent transcriptomic analyses, however, have indicated a significant upregulation of BPGM in acutely injured murine and human kidneys, suggesting a potential renal function for this enzyme. Here we aim to explore the physiological role of BPGM in the kidney. METHODS A tubular-specific, doxycycline-inducible Bpgm-knockout mouse model was generated. Histological, immunofluorescence, and proteomic analyses were conducted to examine the localization of BPGM expression and the impact of its knockout on kidney structure and function. In vitro studies were performed to investigate the metabolic consequences of Bpgm knockdown under osmotic stress. RESULTS BPGM expression was localized to the distal nephron and was absent in proximal tubules. Inducible knockout of Bpgm resulted in rapid kidney injury within 4 days, characterized by proximal tubular damage and tubulointerstitial fibrosis. Proteomic analyses revealed involvement of BPGM in key metabolic pathways, including glycolysis, oxidative stress response, and inflammation. In vitro, Bpgm knockdown led to enhanced glycolysis, decreased reactive oxygen species elimination capacity under osmotic stress, and increased apoptosis. Furthermore, interactions between nephron segments and immune cells in the kidney suggested a mechanism for propagating stress signals from distal to proximal tubules. CONCLUSION BPGM fulfills critical functions beyond the erythrocyte in maintaining glucose metabolism in the distal nephron. Its absence leads to metabolic imbalances, increased oxidative stress, inflammation, and ultimately kidney injury.
Collapse
Affiliation(s)
- Vera A Kulow
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Kameliya Roegner
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Robert Labes
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Mumtaz Kasim
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Susanne Mathia
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Claudia S Czopek
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Nikolaus Berndt
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Deutsches Herzzentrum der Charité (DHZC), Institute of Computer-assisted Cardiovascular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp N Becker
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Gohar Ter-Avetisyan
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Enghard
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Christian Hinze
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jan Klocke
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Kai-Uwe Eckardt
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Kai M Schmidt-Ott
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Pontus B Persson
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| | - Christian Rosenberger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m.S. Nephrologie und Internistische Intensivmedizin (CCM), Berlin, Germany
| | - Michael Fähling
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Translationale Physiologie (CCM), Berlin, Germany
| |
Collapse
|
5
|
Wu J, Guo J, Xia S, Chen J, Cao M, Xie L, Yang C, Qiu F, Wang J. A Single-Cell Transcriptome Profiling of Triptolide-Induced Nephrotoxicity in Mice. Adv Biol (Weinh) 2024; 8:e2400120. [PMID: 38864263 DOI: 10.1002/adbi.202400120] [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: 02/29/2024] [Revised: 05/16/2024] [Indexed: 06/13/2024]
Abstract
Triptolide (TP), an active component isolated from the traditional Chinese herb Tripterygium wilfordii Hook F (TWHF), shows great promise for treating inflammation-related diseases. However, its potential nephrotoxic effects remain concerning. The mechanism underlying TP-induced nephrotoxicity is inadequately elucidated, particularly at single-cell resolution. Hence, single-cell RNA sequencing (scRNA-seq) of kidney tissues from control and TP-treated mice is performed to generate a thorough description of the renal cell atlas upon TP treatment. Heterogeneous responses of nephron epithelial cells are observed after TP exposure, attributing differential susceptibility of cell subtypes to excessive reactive oxygen species and increased inflammatory responses. Moreover, TP disrupts vascular function by activating endothelial cell immunity and damaging fibroblasts. Severe immune cell damage and the activation of pro-inflammatory Macro_C1 cells are also observed with TP treatment. Additionally, ligand-receptor crosstalk analysis reveals that the SPP1 (osteopontin) signaling pathway targeting Macro_C1 cells is triggered by TP treatment, which may promote the infiltration of Macro_C1 cells to exacerbate renal toxicity. Overall, this study provides comprehensive information on the transcriptomic profiles and cellular composition of TP-associated nephrotoxicity at single-cell resolution, which can strengthen the understanding of the pathogenesis of TP-induced nephrotoxicity and provide valuable clues for the discovery of new therapeutic targets to ameliorate TP-associated nephrotoxicity.
Collapse
Affiliation(s)
- Jiangpeng Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Jinan Guo
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Siyu Xia
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Jiayun Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Min Cao
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Lulin Xie
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Chuanbin Yang
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jigang Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Department of Urology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China
| |
Collapse
|
6
|
Border S, Ferreira RM, Lucarelli N, Manthey D, Kumar S, Paul A, Mimar S, Naglah A, Cheng YH, Barisoni L, Ray J, Strekalova Y, Rosenberg AZ, Tomaszewski JE, Hodgin JB, El-Achkar TM, Jain S, Eadon MT, Sarder P. FUSION: A web-based application for in-depth exploration of multi-omics data with brightfield histology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602778. [PMID: 39026885 PMCID: PMC11257503 DOI: 10.1101/2024.07.09.602778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Spatial -OMICS technologies facilitate the interrogation of molecular profiles in the context of the underlying histopathology and tissue microenvironment. Paired analysis of histopathology and molecular data can provide pathologists with otherwise unobtainable insights into biological mechanisms. To connect the disparate molecular and histopathologic features into a single workspace, we developed FUSION (Functional Unit State IdentificatiON in WSIs [Whole Slide Images]), a web-based tool that provides users with a broad array of visualization and analytical tools including deep learning-based algorithms for in-depth interrogation of spatial -OMICS datasets and their associated high-resolution histology images. FUSION enables end-to-end analysis of functional tissue units (FTUs), automatically aggregating underlying molecular data to provide a histopathology-based medium for analyzing healthy and altered cell states and driving new discoveries using "pathomic" features. We demonstrate FUSION using 10x Visium spatial transcriptomics (ST) data from both formalin-fixed paraffin embedded (FFPE) and frozen prepared datasets consisting of healthy and diseased tissue. Through several use-cases, we demonstrate how users can identify spatial linkages between quantitative pathomics, qualitative image characteristics, and spatial --omics.
Collapse
Affiliation(s)
- Samuel Border
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | | | - Nicholas Lucarelli
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | | | - Suhas Kumar
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Anindya Paul
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Sayat Mimar
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Ahmed Naglah
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Ying-Hua Cheng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Laura Barisoni
- Department of Pathology, Division of AI and Computational Pathology, Duke University, Durham, NC
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC
| | - Jessica Ray
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL
| | - Yulia Strekalova
- College of Public Health and Health Professions, University of Florida, Gainesville, FL
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine Baltimore, MD
| | - John E Tomaszewski
- Department of Pathology & Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | | | - Tarek M El-Achkar
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Indianapolis VA Medical Center, Indianapolis, IN
| | - Sanjay Jain
- Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, MO
| | - Michael T Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Pinaki Sarder
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| |
Collapse
|
7
|
O'Hara DV, Lam CSP, McMurray JJV, Yi TW, Hocking S, Dawson J, Raichand S, Januszewski AS, Jardine MJ. Applications of SGLT2 inhibitors beyond glycaemic control. Nat Rev Nephrol 2024; 20:513-529. [PMID: 38671190 DOI: 10.1038/s41581-024-00836-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2024] [Indexed: 04/28/2024]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors were initially developed for their glucose-lowering effects and have shown a modest glycaemic benefit in people with type 2 diabetes mellitus (T2DM). In the past decade, a series of large, robust clinical trials of these therapies have demonstrated striking beneficial effects for various care goals, transforming the chronic disease therapeutic landscape. Cardiovascular safety studies in people with T2DM demonstrated that SGLT2 inhibitors reduce cardiovascular death and hospitalization for heart failure. Subsequent trials in participants with heart failure with reduced or preserved left ventricular ejection fraction demonstrated that SGLT2 inhibitors have beneficial effects on heart failure outcomes. In dedicated kidney outcome studies, SGLT2 inhibitors reduced the incidence of kidney failure among participants with or without diabetes. Post hoc analyses have suggested a range of other benefits of these drugs in conditions as diverse as metabolic dysfunction-associated steatotic liver disease, kidney stone prevention and anaemia. SGLT2 inhibitors have a generally favourable adverse effect profile, although patient selection and medication counselling remain important. Concerted efforts are needed to better integrate these agents into routine care and support long-term medication adherence to close the gap between clinical trial outcomes and those achieved in the real world.
Collapse
Affiliation(s)
- Daniel V O'Hara
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Carolyn S P Lam
- National Heart Centre Singapore, Duke-NUS Medical School, Singapore, Singapore
- Baim Institute for Clinical Research, Boston, MA, USA
| | - John J V McMurray
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Tae Won Yi
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- The George Institute for Global Health, University of New South Wales, Newtown, New South Wales, Australia
| | - Samantha Hocking
- Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
- Boden Initiative, Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Jessica Dawson
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Nutrition and Dietetics, St George Hospital, Kogarah, New South Wales, Australia
| | - Smriti Raichand
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- Centre for the Health Economy (MUCHE), Macquarie University, Macquarie Park, New South Wales, Australia
| | - Andrzej S Januszewski
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Medicine (St. Vincent's Hospital), The University of Melbourne, Fitzroy, Victoria, Australia
- Sydney Pharmacy School, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Meg J Jardine
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia.
- Department of Renal Medicine, Concord Repatriation General Hospital, Concord, New South Wales, Australia.
| |
Collapse
|
8
|
Phillips PCA, de Sousa Loreto Aresta Branco M, Cliff CL, Ward JK, Squires PE, Hills CE. Targeting senescence to prevent diabetic kidney disease: Exploring molecular mechanisms and potential therapeutic targets for disease management. Diabet Med 2024:e15408. [PMID: 38995865 DOI: 10.1111/dme.15408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND/AIMS As a microvascular complication, diabetic kidney disease is the leading cause of chronic kidney disease and end-stage renal disease worldwide. While the underlying pathophysiology driving transition of diabetic kidney disease to renal failure is yet to be fully understood, recent studies suggest that cellular senescence is central in disease development and progression. Consequently, understanding the molecular mechanisms which initiate and drive senescence in response to the diabetic milieu is crucial in developing targeted therapies that halt progression of renal disease. METHODS To understand the mechanistic pathways underpinning cellular senescence in the context of diabetic kidney disease, we reviewed the literature using PubMed for English language articles that contained key words related to senescence, inflammation, fibrosis, senescence-associated secretory phenotype (SASP), autophagy, and diabetes. RESULTS Aberrant accumulation of metabolically active senescent cells is a notable event in the progression of diabetic kidney disease. Through autocrine- and paracrine-mediated mechanisms, resident senescent cells potentiate inflammation and fibrosis through increased expression and secretion of pro-inflammatory cytokines, chemoattractants, recruitment of immune cells, myofibroblast activation, and extracellular matrix remodelling. Compounds that eliminate senescent cells and/or target the SASP - including senolytic and senomorphics drugs - demonstrate promising results in reducing the senescent cell burden and associated pro-inflammatory effect. CONCLUSIONS Here we evidence the link between senescence and diabetic kidney disease and highlight underlying molecular mechanisms and potential therapeutic targets that could be exploited to delay disease progression and improve outcomes for individuals with the disease. Trials are now required to translate their therapeutic potential to a clinical setting.
Collapse
Affiliation(s)
| | | | | | - Joanna Kate Ward
- Joseph Banks Laboratories, College of Health and Science, Lincoln, UK
| | | | | |
Collapse
|
9
|
Musah S, Bhattacharya R, Himmelfarb J. Kidney Disease Modeling with Organoids and Organs-on-Chips. Annu Rev Biomed Eng 2024; 26:383-414. [PMID: 38424088 PMCID: PMC11479997 DOI: 10.1146/annurev-bioeng-072623-044010] [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: 03/02/2024]
Abstract
Kidney disease is a global health crisis affecting more than 850 million people worldwide. In the United States, annual Medicare expenditures for kidney disease and organ failure exceed $81 billion. Efforts to develop targeted therapeutics are limited by a poor understanding of the molecular mechanisms underlying human kidney disease onset and progression. Additionally, 90% of drug candidates fail in human clinical trials, often due to toxicity and efficacy not accurately predicted in animal models. The advent of ex vivo kidney models, such as those engineered from induced pluripotent stem (iPS) cells and organ-on-a-chip (organ-chip) systems, has garnered considerable interest owing to their ability to more accurately model tissue development and patient-specific responses and drug toxicity. This review describes recent advances in developing kidney organoids and organ-chips by harnessing iPS cell biology to model human-specific kidney functions and disease states. We also discuss challenges that must be overcome to realize the potential of organoids and organ-chips as dynamic and functional conduits of the human kidney. Achieving these technological advances could revolutionize personalized medicine applications and therapeutic discovery for kidney disease.
Collapse
Affiliation(s)
- Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina, USA;
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, North Carolina, USA
- Developmental and Stem Cell Biology Program and Department of Cell Biology, Duke University, Durham, North Carolina, USA
| | - Rohan Bhattacharya
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina, USA;
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, North Carolina, USA
| | - Jonathan Himmelfarb
- Department of Medicine, Kidney Research Institute, and Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA;
| |
Collapse
|
10
|
Zhang Y, Bock F, Ferdaus M, Arroyo JP, L Rose K, Patel P, Denton JS, Delpire E, Weinstein AM, Zhang MZ, Harris RC, Terker AS. Low potassium activation of proximal mTOR/AKT signaling is mediated by Kir4.2. Nat Commun 2024; 15:5144. [PMID: 38886379 PMCID: PMC11183202 DOI: 10.1038/s41467-024-49562-w] [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: 08/31/2023] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
The renal epithelium is sensitive to changes in blood potassium (K+). We identify the basolateral K+ channel, Kir4.2, as a mediator of the proximal tubule response to K+ deficiency. Mice lacking Kir4.2 have a compensated baseline phenotype whereby they increase their distal transport burden to maintain homeostasis. Upon dietary K+ depletion, knockout animals decompensate as evidenced by increased urinary K+ excretion and development of a proximal renal tubular acidosis. Potassium wasting is not proximal in origin but is caused by higher ENaC activity and depends upon increased distal sodium delivery. Three-dimensional imaging reveals Kir4.2 knockouts fail to undergo proximal tubule expansion, while the distal convoluted tubule response is exaggerated. AKT signaling mediates the dietary K+ response, which is blunted in Kir4.2 knockouts. Lastly, we demonstrate in isolated tubules that AKT phosphorylation in response to low K+ depends upon mTORC2 activation by secondary changes in Cl- transport. Data support a proximal role for cell Cl- which, as it does along the distal nephron, responds to K+ changes to activate kinase signaling.
Collapse
Affiliation(s)
- Yahua Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Fabian Bock
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Mohammed Ferdaus
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juan Pablo Arroyo
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Kristie L Rose
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Purvi Patel
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weil Medical College, New York, NY, USA
| | - Ming-Zhi Zhang
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
| | - Raymond C Harris
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Andrew S Terker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Kidney Disease, Nashville, TN, USA.
| |
Collapse
|
11
|
Powell NR, Shugg T, Leighty J, Martin M, Kreutz RP, Eadon MT, Lai D, Lu T, Skaar TC. Analysis of the combined effect of rs699 and rs5051 on angiotensinogen expression and hypertension. Chronic Dis Transl Med 2024; 10:102-117. [PMID: 38872760 PMCID: PMC11166681 DOI: 10.1002/cdt3.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 06/15/2024] Open
Abstract
Background Hypertension (HTN) involves genetic variability in the renin-angiotensin system and influences antihypertensive response. We previously reported that angiotensinogen (AGT) messenger RNA (mRNA) is endogenously bound by miR-122-5p and rs699 A > G decreases reporter mRNA in the microRNA functional-assay PASSPORT-seq. The AGT promoter variant rs5051 C > T is in linkage disequilibrium (LD) with rs699 A > G and increases AGT transcription. The independent effect of these variants is understudied due to their LD therefore we aimed to test the hypothesis that increased AGT by rs5051 C > T counterbalances AGT decreased by rs699 A > G, and when these variants occur independently, it translates to HTN-related phenotypes. Methods We used in silico, in vitro, in vivo, and retrospective models to test this hypothesis. Results In silico, rs699 A > G is predicted to increase miR-122-5p binding affinity by 3%. Mir-eCLIP results show rs699 is 40-45 nucleotides from the strongest microRNA-binding site in the AGT mRNA. Unexpectedly, rs699 A > G increases AGT mRNA in an AGT-plasmid-cDNA HepG2 expression model. Genotype-Tissue Expression (GTEx) and UK Biobank analyses demonstrate liver AGT expression and HTN phenotypes are not different when rs699 A > G occurs independently from rs5051 C > T. However, GTEx and the in vitro experiments suggest rs699 A > G confers cell-type-specific effects on AGT mRNA abundance, and suggest paracrine renal renin-angiotensin-system perturbations could mediate the rs699 A > G associations with HTN. Conclusions We found that rs5051 C > T and rs699 A > G significantly associate with systolic blood pressure in Black participants in the UK Biobank, demonstrating a fourfold larger effect than in White participants. Further studies are warranted to determine if altered antihypertensive response in Black individuals might be due to rs5051 C > T or rs699 A > G. Studies like this will help clinicians move beyond the use of race as a surrogate for genotype.
Collapse
Affiliation(s)
- Nicholas R. Powell
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Tyler Shugg
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Jacob Leighty
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Matthew Martin
- Department of Pharmacology and ToxicologySchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Rolf P. Kreutz
- Department of CardiologySchool of Medicine, Krannert Institute of Cardiology, Indiana UniversityIndianapolisIndianaUSA
| | - Michael T. Eadon
- Division of Nephrology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Dongbing Lai
- Department of Medical and Molecular GeneticsSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Tao Lu
- Department of Pharmacology and ToxicologySchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Todd C. Skaar
- Division of Clinical Pharmacology, Department of MedicineSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsSchool of Medicine, Indiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
12
|
Seifi N, Bahari H, Nosrati M, Koochakpoor G, Alizadeh Hassani Z, Rastegarmoghadam-Ebrahimian A, Abedsaeidi M, Ferns GA, Ghyour-Mobarhan M. Higher dietary acid load is associated with the risk of hyperuricemia. Int Urol Nephrol 2024; 56:1743-1749. [PMID: 38072898 DOI: 10.1007/s11255-023-03876-8] [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: 04/28/2023] [Accepted: 11/02/2023] [Indexed: 04/09/2024]
Abstract
PURPOSE Dietary acid load plays a key role in regulating serum uric acid levels. We hypothesized that dietary acid load indices would be positively associated with the odds of hyperuricemia. We aimed to test this hypothesis in a representative sample of Iranian adult population. METHODS In this cross-sectional study, a total of 6145 participants aged 35-65 years were recruited from MASHAD cohort study. Dietary intakes were assessed using a 24-h dietary recall. Diet-based acid load was assessed as the potential renal acid load (PRAL), net endogenous acid production (NEAP), and dietary acid load (DAL). Hyperuricemia was defined as serum uric acid greater than the 75th percentile. Multivariable logistic regression models were applied to determine the association between diet-based acid load scores and hyperuricemia. RESULTS The mean age of participants was 48.89 ± 8.09 years. Overall, 25.7% had hyperuricemia. According to the full-adjusted model, there was a significant association between higher tertile of PRAL, and DAL and hyperuricemia (Q3 PRAL; OR (95% CI): 1.23 (1.05-1.43), Q3 DAL; OR (95% CI): 1.22 (1.05-1.42)). Regarding NEAP, there was no significant association with hyperuricemia. We also found that dietary intake of total sugars, fiber, calcium, and magnesium was associated with the odds of hyperuricemia in our population. CONCLUSION This study showed a significant positive association between two indicators of dietary acid load (PRAL, and DAL) and odds of hyperuricemia among Iranian adults.
Collapse
Affiliation(s)
- Najmeh Seifi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Bahari
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Nosrati
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Glareh Koochakpoor
- School of Nursing and Allied Medical Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Zahra Alizadeh Hassani
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Arezoo Rastegarmoghadam-Ebrahimian
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihehsadat Abedsaeidi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton &Amp, Sussex Medical School, Falmer, Brighton, Sussex, UK
| | - Majid Ghyour-Mobarhan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran.
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Gombedza FC, Shin S, Sadiua J, Stackhouse GB, Bandyopadhyay BC. The Rise in Tubular pH during Hypercalciuria Exacerbates Calcium Stone Formation. Int J Mol Sci 2024; 25:4787. [PMID: 38732005 PMCID: PMC11084476 DOI: 10.3390/ijms25094787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
In calcium nephrolithiasis (CaNL), most calcium kidney stones are identified as calcium oxalate (CaOx) with variable amounts of calcium phosphate (CaP), where CaP is found as the core component. The nucleation of CaP could be the first step of CaP+CaOx (mixed) stone formation. High urinary supersaturation of CaP due to hypercalciuria and an elevated urine pH have been described as the two main factors in the nucleation of CaP crystals. Our previous in vivo findings (in mice) show that transient receptor potential canonical type 3 (TRPC3)-mediated Ca2+ entry triggers a transepithelial Ca2+ flux to regulate proximal tubular (PT) luminal [Ca2+], and TRPC3-knockout (KO; -/-) mice exhibited moderate hypercalciuria and microcrystal formation at the loop of Henle (LOH). Therefore, we utilized TRPC3 KO mice and exposed them to both hypercalciuric [2% calcium gluconate (CaG) treatment] and alkalineuric conditions [0.08% acetazolamide (ACZ) treatment] to generate a CaNL phenotype. Our results revealed a significant CaP and mixed crystal formation in those treated KO mice (KOT) compared to their WT counterparts (WTT). Importantly, prolonged exposure to CaG and ACZ resulted in a further increase in crystal size for both treated groups (WTT and KOT), but the KOT mice crystal sizes were markedly larger. Moreover, kidney tissue sections of the KOT mice displayed a greater CaP and mixed microcrystal formation than the kidney sections of the WTT group, specifically in the outer and inner medullary and calyceal region; thus, a higher degree of calcifications and mixed calcium lithiasis in the kidneys of the KOT group was displayed. In our effort to find the Ca2+ signaling pathophysiology of PT cells, we found that PT cells from both treated groups (WTT and KOT) elicited a larger Ca2+ entry compared to the WT counterparts because of significant inhibition by the store-operated Ca2+ entry (SOCE) inhibitor, Pyr6. In the presence of both SOCE (Pyr6) and ROCE (receptor-operated Ca2+ entry) inhibitors (Pyr10), Ca2+ entry by WTT cells was moderately inhibited, suggesting that the Ca2+ and pH levels exerted sensitivity changes in response to ROCE and SOCE. An assessment of the gene expression profiles in the PT cells of WTT and KOT mice revealed a safeguarding effect of TRPC3 against detrimental processes (calcification, fibrosis, inflammation, and apoptosis) in the presence of higher pH and hypercalciuric conditions in mice. Together, these findings show that compromise in both the ROCE and SOCE mechanisms in the absence of TRPC3 under hypercalciuric plus higher tubular pH conditions results in higher CaP and mixed crystal formation and that TRPC3 is protective against those adverse effects.
Collapse
Affiliation(s)
- Farai C. Gombedza
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; (F.C.G.); (S.S.); (J.S.)
| | - Samuel Shin
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; (F.C.G.); (S.S.); (J.S.)
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC 20064, USA
| | - Jaclyn Sadiua
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; (F.C.G.); (S.S.); (J.S.)
| | - George B. Stackhouse
- Urology Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA;
| | - Bidhan C. Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; (F.C.G.); (S.S.); (J.S.)
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC 20064, USA
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
14
|
Walker V. The Intricacies of Renal Phosphate Reabsorption-An Overview. Int J Mol Sci 2024; 25:4684. [PMID: 38731904 PMCID: PMC11083860 DOI: 10.3390/ijms25094684] [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: 03/24/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
To maintain an optimal body content of phosphorus throughout postnatal life, variable phosphate absorption from food must be finely matched with urinary excretion. This amazing feat is accomplished through synchronised phosphate transport by myriads of ciliated cells lining the renal proximal tubules. These respond in real time to changes in phosphate and composition of the renal filtrate and to hormonal instructions. How they do this has stimulated decades of research. New analytical techniques, coupled with incredible advances in computer technology, have opened new avenues for investigation at a sub-cellular level. There has been a surge of research into different aspects of the process. These have verified long-held beliefs and are also dramatically extending our vision of the intense, integrated, intracellular activity which mediates phosphate absorption. Already, some have indicated new approaches for pharmacological intervention to regulate phosphate in common conditions, including chronic renal failure and osteoporosis, as well as rare inherited biochemical disorders. It is a rapidly evolving field. The aim here is to provide an overview of our current knowledge, to show where it is leading, and where there are uncertainties. Hopefully, this will raise questions and stimulate new ideas for further research.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton S016 6YD, UK
| |
Collapse
|
15
|
Chambers BE, Weaver NE, Lara CM, Nguyen TK, Wingert RA. (Zebra)fishing for nephrogenesis genes. Tissue Barriers 2024; 12:2219605. [PMID: 37254823 PMCID: PMC11042071 DOI: 10.1080/21688370.2023.2219605] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/14/2023] [Indexed: 06/01/2023] Open
Abstract
Kidney disease is a devastating condition affecting millions of people worldwide, where over 100,000 patients in the United States alone remain waiting for a lifesaving organ transplant. Concomitant with a surge in personalized medicine, single-gene mutations, and polygenic risk alleles have been brought to the forefront as core causes of a spectrum of renal disorders. With the increasing prevalence of kidney disease, it is imperative to make substantial strides in the field of kidney genetics. Nephrons, the core functional units of the kidney, are epithelial tubules that act as gatekeepers of body homeostasis by absorbing and secreting ions, water, and small molecules to filter the blood. Each nephron contains a series of proximal and distal segments with explicit metabolic functions. The embryonic zebrafish provides an ideal platform to systematically dissect the genetic cues governing kidney development. Here, we review the use of zebrafish to discover nephrogenesis genes.
Collapse
Affiliation(s)
- Brooke E. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Nicole E. Weaver
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Caroline M. Lara
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| |
Collapse
|
16
|
Shaked SA, Abehsera S, Ziegler A, Bentov S, Manor R, Weil S, Ohana E, Eichler J, Aflalo ED, Sagi A. A transporter that allows phosphate ions to control the polymorph of exoskeletal calcium carbonate biomineralization. Acta Biomater 2024; 178:221-232. [PMID: 38428510 DOI: 10.1016/j.actbio.2024.02.035] [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: 10/15/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
The SLC20A2 transporter supplies phosphate ions (Pi) for diverse biological functions in vertebrates, yet has not been studied in crustaceans. Unlike vertebrates, whose skeletons are mineralized mainly by calcium phosphate, only minute amounts of Pi are found in the CaCO3-mineralized exoskeletons of invertebrates. In this study, a crustacean SLC20A2 transporter was discovered and Pi transport to exoskeletal elements was studied with respect to the role of Pi in invertebrate exoskeleton biomineralization, revealing an evolutionarily conserved mechanism for Pi transport in both vertebrates and invertebrates. Freshwater crayfish, including the study animal Cherax quadricarinatus, require repeated molt cycles for their growth. During the molt cycle, crayfish form transient exoskeletal mineral storage organs named gastroliths, which mostly contain amorphous calcium carbonate (ACC), an unstable polymorph long-thought to be stabilized by Pi. RNA interference experiments via CqSLC20A2 dsRNA injections reduced Pi content in C. quadricarinatus gastroliths, resulting in increased calcium carbonate (CaCO3) crystallinity and grain size. The discovery of a SLC20A2 transporter in crustaceans and the demonstration that knocking down its mRNA reduced Pi content in exoskeletal elements offers the first direct proof of a long-hypothesized mechanism by which Pi affects CaCO3 biomineralization in the crustacean exoskeleton. This research thus demonstrated the distinct role of Pi as an amorphous mineral polymorph stabilizer in vivo, suggesting further avenues for amorphous biomaterial studies. STATEMENT OF SIGNIFICANCE: • Crustaceans exoskeletons are hardened mainly by CaCO3, with Pi in minute amounts • Pi was hypothesized to stabilize exoskeletal amorphous mineral forms in vivo • For the first time, transport protein for Pi was discovered in crayfish • Transport knock-down resulted in exoskeletal CaCO3 crystallization and reduced Pi.
Collapse
Affiliation(s)
- Shai A Shaked
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Shai Abehsera
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Andreas Ziegler
- Central Facility for Electron Microscopy, University of Ulm, Albert-Einstein-Allee 11, 89069 Ulm, Germany
| | - Shmuel Bentov
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Rivka Manor
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Simy Weil
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ehud Ohana
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jerry Eichler
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Eliahu D Aflalo
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; Department of Life Sciences, Achva Academic College, 79804, Israel
| | - Amir Sagi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| |
Collapse
|
17
|
Kuhn C, Mohebbi N, Ritter A. Metabolic acidosis in chronic kidney disease: mere consequence or also culprit? Pflugers Arch 2024; 476:579-592. [PMID: 38279993 PMCID: PMC11006741 DOI: 10.1007/s00424-024-02912-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/29/2024]
Abstract
Metabolic acidosis is a frequent complication in non-transplant chronic kidney disease (CKD) and after kidney transplantation. It occurs when net endogenous acid production exceeds net acid excretion. While nephron loss with reduced ammoniagenesis is the main cause of acid retention in non-transplant CKD patients, additional pathophysiological mechanisms are likely inflicted in kidney transplant recipients. Functional tubular damage by calcineurin inhibitors seems to play a key role causing renal tubular acidosis. Notably, experimental and clinical studies over the past decades have provided evidence that metabolic acidosis may not only be a consequence of CKD but also a driver of disease. In metabolic acidosis, activation of hormonal systems and the complement system resulting in fibrosis have been described. Further studies of changes in renal metabolism will likely contribute to a deeper understanding of the pathophysiology of metabolic acidosis in CKD. While alkali supplementation in case of reduced serum bicarbonate < 22 mmol/l has been endorsed by CKD guidelines for many years to slow renal functional decline, among other considerations, beneficial effects and thresholds for treatment have lately been under intense debate. This review article discusses this topic in light of the most recent results of trials assessing the efficacy of dietary and pharmacological interventions in CKD and kidney transplant patients.
Collapse
Affiliation(s)
- Christian Kuhn
- Clinic for Nephrology and Transplantation Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | | | - Alexander Ritter
- Clinic for Nephrology and Transplantation Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland.
- Clinic for Nephrology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Roskosch J, Huynh-Do U, Rudloff S. Lectin-mediated, time-efficient, and high-yield sorting of different morphologically intact nephron segments. Pflugers Arch 2024; 476:379-393. [PMID: 38091061 PMCID: PMC10847228 DOI: 10.1007/s00424-023-02894-w] [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: 02/28/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 02/08/2024]
Abstract
The kidney is a highly complex organ equipped with a multitude of miniscule filter-tubule units called nephrons. Each nephron can be subdivided into multiple segments, each with its own morphology and physiological function. To date, conventional manual approaches to isolate specific nephron segments are very laborious, time-consuming, often limited to only a specific segment, and typically have low yield. Here, we describe a novel, unconventional method that is superior in many aspects to previous protocols by combining low-cost fluorophore-conjugated lectins or agglutinins (Flaggs) with flow sorting. This allows the simultaneous separation of different nephron segments with preserved 3D morphology from mouse or human samples in under 3 h. Using a 200-µm nozzle and 5 psi, glomeruli, proximal, or distal convoluted tubules are sorted with Cy3-labeled Sambucus Nigra agglutinin (SNA-Cy3), Fluorescein-labeled Lotus Tetragonolobus lectin (LTL-FITC), or Pacific Blue-labeled soybean agglutinin (SBA-PB), respectively. Connecting tubules and collecting ducts are sorted by double-positive SBA-PB and SNA-Cy3 signals, while thick ascending limb segments are characterized by the absence of any Flaggs labeling. From two mouse kidneys, this yields 37-521 ng protein/s or 0.71-16.71 ng RNA/s, depending on the specific nephron segment. The purity of sorted segments, as assessed by mRNA expression level profiling of 15 genes, is very high with a 96.1-fold median enrichment across all genes and sorted segments. In summary, our method represents a simple, straightforward, cost-effective, and widely applicable tool yielding high amounts of pure and morphologically largely intact renal tubule materials with the potential to propel nephron segment-specific research.
Collapse
Affiliation(s)
- Jessica Roskosch
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Uyen Huynh-Do
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Stefan Rudloff
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland.
| |
Collapse
|
19
|
Chrysopoulou M, Rinschen MM. Metabolic Rewiring and Communication: An Integrative View of Kidney Proximal Tubule Function. Annu Rev Physiol 2024; 86:405-427. [PMID: 38012048 DOI: 10.1146/annurev-physiol-042222-024724] [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: 11/29/2023]
Abstract
The kidney proximal tubule is a key organ for human metabolism. The kidney responds to stress with altered metabolite transformation and perturbed metabolic pathways, an ultimate cause for kidney disease. Here, we review the proximal tubule's metabolic function through an integrative view of transport, metabolism, and function, and embed it in the context of metabolome-wide data-driven research. Function (filtration, transport, secretion, and reabsorption), metabolite transformation, and metabolite signaling determine kidney metabolic rewiring in disease. Energy metabolism and substrates for key metabolic pathways are orchestrated by metabolite sensors. Given the importance of renal function for the inner milieu, we also review metabolic communication routes with other organs. Exciting research opportunities exist to understand metabolic perturbation of kidney and proximal tubule function, for example, in hypertension-associated kidney disease. We argue that, based on the integrative view outlined here, kidney diseases without genetic cause should be approached scientifically as metabolic diseases.
Collapse
Affiliation(s)
| | - Markus M Rinschen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark;
- III. Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| |
Collapse
|
20
|
Yolanda H, Jearawuttanakul K, Wannalo W, Kanjanasirirat P, Borwornpinyo S, Rujirawat T, Payattikul P, Kittichotirat W, Wichadakul D, Krajaejun T. Potential anti- Pythium insidiosum therapeutics identified through screening of agricultural fungicides. Microbiol Spectr 2024; 12:e0162023. [PMID: 38179943 PMCID: PMC10846074 DOI: 10.1128/spectrum.01620-23] [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: 07/07/2023] [Indexed: 01/06/2024] Open
Abstract
Pythiosis is a life-threatening infectious disease caused by the oomycete Pythium insidiosum. Clinical manifestations of pythiosis include an eye, blood vessel, skin, or gastrointestinal tract infection. Pythiosis has been increasingly reported worldwide, with an overall mortality rate of 28%. Radical surgery is required to save patients' lives due to the limited efficacy of antimicrobial drugs. Effective medical treatments are urgently needed for pythiosis. This study aims to find anti-P. insidiosum agents by screening 17 agricultural fungicides that inhibit plant-pathogenic oomycetes and validating their efficacy and safety. Cyazofamid outperformed other fungicides as it can potently inhibit genetically diverse P. insidiosum isolates while exhibiting minimal cellular toxicities. The calculated therapeutic scores determined that the concentration of cyazofamid causing significant cellular toxicities was eight times greater than the concentration of the drug effectively inhibiting P. insidiosum. Furthermore, other studies showed that cyazofamid exhibits low-to-moderate toxicities in animals. The mechanism of cyazofamid action is likely the inhibition of cytochrome b, an essential component in ATP synthesis. Molecular docking and dynamic analyses depicted a stable binding of cyazofamid to the Qi site of the P. insidiosum's cytochrome b orthologous protein. In conclusion, our search for an effective anti-P. insidiosum drug indicated that cyazofamid is a promising candidate for treating pythiosis. With its high efficacy and low toxicity, cyazofamid is a potential chemical for treating pythiosis, reducing the need for radical surgeries, and improving recovery rates. Our findings could pave the way for the development of new and effective treatments for pythiosis.IMPORTANCEPythiosis is a severe infection caused by Pythium insidiosum. The disease is prevalent in tropical/subtropical regions. This infectious condition is challenging to treat with antifungal drugs and often requires surgical removal of the infected tissue. Pythiosis can be fatal if not treated promptly. There is a need for a new treatment that effectively inhibits P. insidiosum. This study screened 17 agricultural fungicides that target plant-pathogenic oomycetes and found that cyazofamid was the most potent in inhibiting P. insidiosum. Cyazofamid showed low toxicity to mammalian cells and high affinity to the P. insidiosum's cytochrome b, which is involved in energy production. Cyazofamid could be a promising candidate for the treatment of pythiosis, as it could reduce the need for surgery and improve the survival rate of patients. This study provides valuable insights into the biology and drug susceptibility of P. insidiosum and opens new avenues for developing effective therapies for pythiosis.
Collapse
Affiliation(s)
- Hanna Yolanda
- Program in Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Department of Parasitology, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Kedchin Jearawuttanakul
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Warawuth Wannalo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Thidarat Rujirawat
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Penpan Payattikul
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bangkhuntien, Bangkok, Thailand
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bangkhuntien, Bangkok, Thailand
| | - Duangdao Wichadakul
- Department of Computer Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Theerapong Krajaejun
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
21
|
Pasupulati AK, Nagati V, Paturi ASV, Reddy GB. Non-enzymatic glycation and diabetic kidney disease. VITAMINS AND HORMONES 2024; 125:251-285. [PMID: 38997166 DOI: 10.1016/bs.vh.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Chronic diabetes leads to various complications including diabetic kidney disease (DKD). DKD is a major microvascular complication and the leading cause of morbidity and mortality in diabetic patients. Varying degrees of proteinuria and reduced glomerular filtration rate are the cardinal clinical manifestations of DKD that eventually progress into end-stage renal disease. Histopathologically, DKD is characterized by renal hypertrophy, mesangial expansion, podocyte injury, glomerulosclerosis, and tubulointerstitial fibrosis, ultimately leading to renal replacement therapy. Amongst the many mechanisms, hyperglycemia contributes to the pathogenesis of DKD via a mechanism known as non-enzymatic glycation (NEG). NEG is the irreversible conjugation of reducing sugars onto a free amino group of proteins by a series of events, resulting in the formation of initial Schiff's base and an Amadori product and to a variety of advanced glycation end products (AGEs). AGEs interact with cognate receptors and evoke aberrant signaling cascades that execute adverse events such as oxidative stress, inflammation, phenotypic switch, complement activation, and cell death in different kidney cells. Elevated levels of AGEs and their receptors were associated with clinical and morphological manifestations of DKD. In this chapter, we discussed the mechanism of AGEs accumulation, AGEs-induced cellular and molecular events in the kidney and their impact on the pathogenesis of DKD. We have also reflected upon the possible options to curtail the AGEs accumulation and approaches to prevent AGEs mediated adverse renal outcomes.
Collapse
Affiliation(s)
- Anil K Pasupulati
- Department of Biochemistry, University of Hyderabad, Hyderabad, India.
| | - Veerababu Nagati
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
| | - Atreya S V Paturi
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
| | - G Bhanuprakash Reddy
- Department of Biochemistry, ICMR-National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
22
|
Streng KW, Hillege HL, Ter Maaten JM, van Veldhuisen DJ, Dickstein K, Samani NJ, Ng LL, Metra M, Filippatos GS, Ponikowski P, Zannad F, Anker SD, van der Meer P, Lang CC, Voors AA, Damman K. Urinary Marker Profiles in Heart Failure with Reduced Versus Preserved Ejection Fraction. J Cardiovasc Transl Res 2024; 17:3-12. [PMID: 36795286 PMCID: PMC10896953 DOI: 10.1007/s12265-023-10356-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Recent data suggest different causes of renal dysfunction between heart failure with reduced (HFrEF) versus preserved ejection fraction (HFpEF). We therefore studied a wide range of urinary markers reflecting different nephron segments in heart failure patients. METHODS In 2070, in chronic heart failure patients, we measured several established and upcoming urinary markers reflecting different nephron segments. RESULTS Mean age was 70 ± 12 years, 74% was male and 81% (n = 1677) had HFrEF. Mean estimated glomerular filtration rate (eGFR) was lower in patients with HFpEF (56 ± 23 versus 63 ± 23 ml/min/1.73 m2, P = 0.001). Patients with HFpEF had significantly higher values of NGAL (58.1 [24.0-124.8] versus 28.1 [14.6-66.9] μg/gCr, P < 0.001) and KIM-1 (2.28 [1.49-4.37] versus 1.79 [0.85-3.49] μg/gCr, P = 0.001). These differences were more pronounced in patients with an eGFR > 60 ml/min/1.73m2. CONCLUSIONS HFpEF patients showed more evidence of tubular damage and/or dysfunction compared with HFrEF patients, in particular when glomerular function was preserved.
Collapse
Affiliation(s)
- Koen W Streng
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Hans L Hillege
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Jozine M Ter Maaten
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Kenneth Dickstein
- University of Bergen, 5007, Bergen, Norway
- Stavanger University Hospital, Stavanger, Norway
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Marco Metra
- Institute of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Gerasimos S Filippatos
- Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
- University of Cyprus, Nicosia, Cyprus
| | - Piotr Ponikowski
- Department of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
- Cardiology Department, Military Hospital, Wroclaw, Poland
| | - Faiez Zannad
- INSERM, Centre d'Investigations Cliniques Plurithe´Matique 1433, F-CRIN INI-CRCT, INSERM U1116, Universite´ de Lorraine, CHRU de Nancy, Nancy, France
| | - Stefan D Anker
- Division of Cardiology and Metabolism, Department of Cardiology (CVK) and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Peter van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Chim C Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, DD1 9SY, Dundee, Scotland
| | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.
| | - Kevin Damman
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
23
|
Rao VS, Ivey-Miranda JB, Cox ZL, Moreno-Villagomez J, Maulion C, Bellumkonda L, Chang J, Field MP, Wiederin DR, Butler J, Collins SP, Turner JM, Wilson FP, Inzucchi SE, Wilcox CS, Ellison DH, Testani JM. Empagliflozin in Heart Failure: Regional Nephron Sodium Handling Effects. J Am Soc Nephrol 2024; 35:189-201. [PMID: 38073038 PMCID: PMC10843196 DOI: 10.1681/asn.0000000000000269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/25/2023] [Indexed: 02/02/2024] Open
Abstract
SIGNIFICANCE STATEMENT The effect of sodium-glucose cotransporter-2 inhibitors (SGLT2i) on regional tubular sodium handling is poorly understood in humans. In this study, empagliflozin substantially decreased lithium reabsorption in the proximal tubule (PT) (a marker of proximal tubular sodium reabsorption), a magnitude out of proportion to that expected with only inhibition of sodium-glucose cotransporter-2. This finding was not driven by an "osmotic diuretic" effect; however, several parameters changed in a manner consistent with inhibition of the sodium-hydrogen exchanger 3. The large changes in proximal tubular handling were acutely buffered by increased reabsorption in both the loop of Henle and the distal nephron, resulting in the observed modest acute natriuresis with these agents. After 14 days of empagliflozin, natriuresis waned due to increased reabsorption in the PT and/or loop of Henle. These findings confirm in humans that SGLT2i have complex and important effects on renal tubular solute handling. BACKGROUND The effect of SGLT2i on regional tubular sodium handling is poorly understood in humans but may be important for the cardiorenal benefits. METHODS This study used a previously reported randomized, placebo-controlled crossover study of empagliflozin 10 mg daily in patients with diabetes and heart failure. Sodium handling in the PT, loop of Henle (loop), and distal nephron was assessed at baseline and day 14 using fractional excretion of lithium (FELi), capturing PT/loop sodium reabsorption. Assessments were made with and without antagonism of sodium reabsorption through the loop using bumetanide. RESULTS Empagliflozin resulted in a large decrease in sodium reabsorption in the PT (increase in FELi=7.5%±10.6%, P = 0.001), with several observations suggesting inhibition of PT sodium hydrogen exchanger 3. In the absence of renal compensation, this would be expected to result in approximately 40 g of sodium excretion/24 hours with normal kidney function. However, rapid tubular compensation occurred with increased sodium reabsorption both in the loop ( P < 0.001) and distal nephron ( P < 0.001). Inhibition of sodium-glucose cotransporter-2 did not attenuate over 14 days of empagliflozin ( P = 0.14). However, there were significant reductions in FELi ( P = 0.009), fractional excretion of sodium ( P = 0.004), and absolute fractional distal sodium reabsorption ( P = 0.036), indicating that chronic adaptation to SGLT2i results primarily from increased reabsorption in the loop and/or PT. CONCLUSIONS Empagliflozin caused substantial redistribution of intrarenal sodium delivery and reabsorption, providing mechanistic substrate to explain some of the benefits of this class. Importantly, the large increase in sodium exit from the PT was balanced by distal compensation, consistent with SGLT2i excellent safety profile. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER ClinicalTrials.gov ( NCT03027960 ).
Collapse
Affiliation(s)
- Veena S. Rao
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Juan B. Ivey-Miranda
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Hospital de Cardiologia, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Zachary L. Cox
- Department of Pharmacy Practice, Lipscomb University College of Pharmacy, Nashville, Tennessee
- Department of Pharmacy, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Julieta Moreno-Villagomez
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Christopher Maulion
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Lavanya Bellumkonda
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - John Chang
- Section of General Internal Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | | | | | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, Texas
| | - Sean P. Collins
- Department of Emergency Medicine, Geriatric Research, Education and Clinical Center (GRECC), Vanderbilt University Medical Center and Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Jeffrey M. Turner
- Division of Nephrology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - F. Perry Wilson
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Clinical and Translational Research Accelerator, Yale University School of Medicine, New Haven, Connecticut
| | - Silvio E. Inzucchi
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher S. Wilcox
- Division of Nephrology and Hypertension Center, Georgetown University, Washington, DC
| | - David H. Ellison
- Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, Oregon
| | - Jeffrey M. Testani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
24
|
Korkola NC, Stillman MJ. Structural motifs in the early metallation steps of Zn(II) and Cd(II) binding to apo-metallothionein 1a. J Inorg Biochem 2024; 251:112429. [PMID: 38000179 DOI: 10.1016/j.jinorgbio.2023.112429] [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: 09/19/2023] [Revised: 10/26/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023]
Abstract
Many proteins require a metal cofactor to function and these metals are often involved in the protein folding process. The protein metallothionein (MT) has a dynamic structure capable of binding to a variety of metals with different stoichiometries. The most well-understood structure is the seven-metal, two domain structure formed upon metallation using Zn(II) or Cd(II). However, the partially metallated states and the pathways to form these clusters are less well-understood, although it is known that the pathways are pH dependent. Using stopped flow methods, it is shown that the metallation rates of the less cooperative Zn(II) binding pathway is much more impacted by low pH conditions that that of the more cooperative Cd(II) binding pathway. Electrospray ionization mass spectrometry (ESI-MS) methods reveal specific mixtures of bridging and terminally bound MxSy structures form in the first few metallation steps. Using a combination of methods, the data show that the result of unfolding this intrinsically disordered apo-MT structure using guanidinium chloride is that the formation of preliminary bridging structures that form in the first few metallation steps is impeded. The data show that more terminally bound structures form. Our conclusion is that the compact conformation of the native apo-MT at physiological pH allows for rapid formation of complex metal-thiolate structures with high affinity that provides protection from oxidation, a function that is suppressed upon unfolding. Overall, these results highlight both the importance of the apo-MT structure in the metallation pathway, but also the differences in Zn(II) and Cd(II) binding under different conditions.
Collapse
Affiliation(s)
- Natalie C Korkola
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, ON N6A5B7, Canada
| | - Martin J Stillman
- Department of Chemistry, The University of Western Ontario, 1151 Richmond St., London, ON N6A5B7, Canada.
| |
Collapse
|
25
|
Porter A, Vorndran HE, Marciszyn A, Mutchler SM, Subramanya AR, Kleyman TR, Hendershot LM, Brodsky JL, Buck TM. Excess dietary sodium partially restores salt and water homeostasis caused by loss of the endoplasmic reticulum molecular chaperone, GRP170, in the mouse nephron. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.13.575426. [PMID: 38260467 PMCID: PMC10802592 DOI: 10.1101/2024.01.13.575426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The maintenance of fluid and electrolyte homeostasis by the kidney requires proper folding and trafficking of ion channels and transporters in kidney epithelia. Each of these processes requires a specific subset of a diverse class of proteins termed molecular chaperones. One such chaperone is GRP170, which is an Hsp70-like, endoplasmic reticulum (ER)-localized chaperone that plays roles in protein quality control and protein folding in the ER. We previously determined that loss of GRP170 in the mouse nephron leads to hypovolemia, electrolyte imbalance, and rapid weight loss. In addition, GRP170-deficient mice develop an AKI-like phenotype, typified by tubular injury, elevation of clinical kidney injury markers, and induction of the unfolded protein response (UPR). By using an inducible GRP170 knockout cellular model, we confirmed that GRP170 depletion induces the UPR, triggers an apoptotic response, and disrupts protein homeostasis. Based on these data, we hypothesized that UPR induction underlies hyponatremia and volume depletion in rodents, but that these and other phenotypes might be rectified by supplementation with high salt. To test this hypothesis, control and GRP170 tubule-specific knockout mice were provided with a diet containing 8% sodium chloride. We discovered that sodium supplementation improved electrolyte imbalance and reduced clinical kidney injury markers, but was unable to restore weight or tubule integrity. These results are consistent with UPR induction contributing to the kidney injury phenotype in the nephron-specific GR170 knockout model, and that the role of GRP170 in kidney epithelia is essential to both maintain electrolyte balance and cellular protein homeostasis.
Collapse
Affiliation(s)
- Aidan Porter
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
- Division of Pediatric Nephrology, University of Pittsburgh, Pittsburgh, PA
| | - Hannah E. Vorndran
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Allison Marciszyn
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Stephanie M. Mutchler
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Arohan R. Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA
| | - Linda M. Hendershot
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 30105
| | - Jeffrey L. Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Teresa M. Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
26
|
McDonough AA, Harris AN, Xiong LI, Layton AT. Sex differences in renal transporters: assessment and functional consequences. Nat Rev Nephrol 2024; 20:21-36. [PMID: 37684523 PMCID: PMC11090267 DOI: 10.1038/s41581-023-00757-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 09/10/2023]
Abstract
Mammalian kidneys are specialized to maintain fluid and electrolyte homeostasis. The epithelial transport processes along the renal tubule that match output to input have long been the subject of experimental and theoretical study. However, emerging data have identified a new dimension of investigation: sex. Like most tissues, the structure and function of the kidney is regulated by sex hormones and chromosomes. Available data demonstrate sex differences in the abundance of kidney solute and electrolyte transporters, establishing that renal tubular organization and operation are distinctly different in females and males. Newer studies have provided insights into the physiological consequences of these sex differences. Computational simulations predict that sex differences in transporter abundance are likely driven to optimize reproduction, enabling adaptive responses to the nutritional requirements of serial pregnancies and lactation - normal life-cycle changes that challenge the ability of renal transporters to maintain fluid and electrolyte homeostasis. Later in life, females may also undergo menopause, which is associated with changes in disease risk. Although numerous knowledge gaps remain, ongoing studies will provide further insights into the sex-specific mechanisms of sodium, potassium, acid-base and volume physiology throughout the life cycle, which may lead to therapeutic opportunities.
Collapse
Affiliation(s)
- Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| | - Autumn N Harris
- Department of Small Animal Clinical Sciences, University of Florida, College of Veterinary Medicine, Gainesville, FL, USA
| | - Lingyun Ivy Xiong
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Anita T Layton
- Departments of Applied Mathematics and Biology, University of Waterloo, Waterloo, Ontario, Canada
- Cheriton School of Computer Science, University of Waterloo, Waterloo, Ontario, Canada
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
27
|
Kang MJ, Ioannou S, Lougheide Q, Dittmar M, Hsu Y, Pastor-Soler NM. The study of intercalated cells using ex vivo techniques: primary cell culture, cell lines, kidney slices, and organoids. Am J Physiol Cell Physiol 2024; 326:C229-C251. [PMID: 37899748 DOI: 10.1152/ajpcell.00479.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 10/31/2023]
Abstract
This review summarizes methods to study kidney intercalated cell (IC) function ex vivo. While important for acid-base homeostasis, IC dysfunction is often not recognized clinically until it becomes severe. The advantage of using ex vivo techniques is that they allow for the differential evaluation of IC function in controlled environments. Although in vitro kidney tubular perfusion is a classical ex vivo technique to study IC, here we concentrate on primary cell cultures, immortalized cell lines, and ex vivo kidney slices. Ex vivo techniques are useful in evaluating IC signaling pathways that allow rapid responses to extracellular changes in pH, CO2, and bicarbonate (HCO3-). However, these methods for IC work can also be challenging, as cell lines that recapitulate IC do not proliferate easily in culture. Moreover, a "pure" IC population in culture does not necessarily replicate its collecting duct (CD) environment, where ICs are surrounded by the more abundant principal cells (PCs). It is reassuring that many findings obtained in ex vivo IC systems signaling have been largely confirmed in vivo. Some of these newly identified signaling pathways reveal that ICs are important for regulating NaCl reabsorption, thus suggesting new frontiers to target antihypertensive treatments. Moreover, recent single-cell characterization studies of kidney epithelial cells revealed a dual developmental origin of IC, as well as the presence of novel CD cell types with certain IC characteristics. These exciting findings present new opportunities for the study of IC ex vivo and will likely rediscover the importance of available tools in this field.NEW & NOTEWORTHY The study of kidney intercalated cells has been limited by current cell culture and kidney tissue isolation techniques. This review is to be used as a reference to select ex vivo techniques to study intercalated cells. We focused on the use of cell lines and kidney slices as potential useful models to study membrane transport proteins. We also review how novel collecting duct organoids may help better elucidate the role of these intriguing cells.
Collapse
Affiliation(s)
- Min Ju Kang
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Silvia Ioannou
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Quinn Lougheide
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Michael Dittmar
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Young Hsu
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Nuria M Pastor-Soler
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| |
Collapse
|
28
|
Al-Beltagi M, Saeed NK, Bediwy AS, Elbeltagi R, Hasan S, Hamza MB. Renal calcification in children with renal tubular acidosis: What a paediatrician should know. World J Clin Pediatr 2023; 12:295-309. [PMID: 38178934 PMCID: PMC10762599 DOI: 10.5409/wjcp.v12.i5.295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/15/2023] [Accepted: 10/16/2023] [Indexed: 12/08/2023] Open
Abstract
Renal tubular acidosis (RTA) can lead to renal calcification in children, which can cause various complications and impair renal function. This review provides pediatricians with a comprehensive understanding of the relationship between RTA and renal calcification, highlighting essential aspects for clinical management. The article analyzed relevant studies to explore the prevalence, risk factors, underlying mechanisms, and clinical implications of renal calcification in children with RTA. Results show that distal RTA (type 1) is particularly associated with nephrocalcinosis, which presents a higher risk of renal calcification. However, there are limitations to the existing literature, including a small number of studies, heterogeneity in methodologies, and potential publication bias. Longitudinal data and control groups are also lacking, which limits our understanding of long-term outcomes and optimal management strategies for children with RTA and renal calcification. Pediatricians play a crucial role in the early diagnosis and management of RTA to mitigate the risk of renal calcification and associated complications. In addition, alkaline therapy remains a cornerstone in the treatment of RTA, aimed at correcting the acid-base imbalance and reducing the formation of kidney stones. Therefore, early diagnosis and appropriate therapeutic interventions are paramount in preventing and managing renal calcification to preserve renal function and improve long-term outcomes for affected children. Further research with larger sample sizes and rigorous methodologies is needed to optimize the clinical approach to renal calcification in the context of RTA in the pediatric population.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatrics, University Medical Center, King Abdulla Medical City, Dr. Sulaiman Al Habib Medical Group, Manama, Bahrain, Manama 26671, Manama, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Manama, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Bahrain, Busaiteen 15503, Muharraq, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
- Department of Chest Disease, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama, Manama 26671, Manama, Bahrain
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland - Bahrain, Busiateen 15503, Muharraq, Bahrain
| | - Samir Hasan
- Department of Pediatrics, Faculty of Medicine, Tanta University Hospital, Tanta 31511, Algharbia, Egypt
| | - Mohamed Basiony Hamza
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Algharbia, Egypt
| |
Collapse
|
29
|
Almazmomi MA, Esmat A, Naeem A. Acute Kidney Injury: Definition, Management, and Promising Therapeutic Target. Cureus 2023; 15:e51228. [PMID: 38283512 PMCID: PMC10821757 DOI: 10.7759/cureus.51228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Acute kidney injury (AKI) is caused by a sudden loss of renal function, resulting in the build-up of waste products and a significant increase in mortality and morbidity. It is commonly diagnosed in critically ill patients, with its occurrence estimated at up to 50% in patients hospitalized in the intensive critical unit. Despite ongoing efforts, the death rate associated with AKI has remained high over the past half-century. Thus, it is critical to investigate novel therapy options for preventing the epidemic. Many studies have found that inflammation and Toll-like receptor-4 (TLR-4) activation have a significant role in the pathogenesis of AKI. Noteworthy, challenges in the search for efficient pharmacological therapy for AKI have arisen due to the multifaceted origin and complexity of the clinical history of people with the disease. This article focuses on kidney injury's epidemiology, risk factors, and pathophysiological processes. Specifically, it focuses on the role of TLRs especially type 4 in disease development.
Collapse
Affiliation(s)
- Meaad A Almazmomi
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Ahmed Esmat
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Anjum Naeem
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
| |
Collapse
|
30
|
Goto S, Yoshida Y, Hosojima M, Kuwahara S, Kabasawa H, Aoki H, Iida T, Sawada R, Ugamura D, Yoshizawa Y, Takemoto K, Komochi K, Kobayashi R, Kaseda R, Yaoita E, Nagatoishi S, Narita I, Tsumoto K, Saito A. Megalin is involved in angiotensinogen-induced, angiotensin II-mediated ERK1/2 signaling to activate Na + -H + exchanger 3 in proximal tubules. J Hypertens 2023; 41:1831-1843. [PMID: 37682076 DOI: 10.1097/hjh.0000000000003555] [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: 09/09/2023]
Abstract
BACKGROUND Kidney angiotensin (Ang) II is produced mainly from liver-derived, glomerular-filtered angiotensinogen (AGT). Podocyte injury has been reported to increase the kidney Ang II content and induce Na + retention depending on the function of megalin, a proximal tubular endocytosis receptor. However, how megalin regulates the renal content and action of Ang II remains elusive. METHODS We used a mass spectrometry-based, parallel reaction-monitoring assay to quantitate Ang II in plasma, urine, and kidney homogenate of kidney-specific conditional megalin knockout (MegKO) and control (Ctl) mice. We also evaluated the pathophysiological changes in both mouse genotypes under the basal condition and under the condition of increased glomerular filtration of AGT induced by administration of recombinant mouse AGT (rec-mAGT). RESULTS Under the basal condition, plasma and kidney Ang II levels were comparable in the two mouse groups. Ang II was detected abundantly in fresh spot urine in conditional MegKO mice. Megalin was also found to mediate the uptake of intravenously administered fluorescent Ang II by PTECs. Administration of rec-mAGT increased kidney Ang II, exerted renal extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, activated proximal tubular Na + -H + exchanger 3 (NHE3), and decreased urinary Na + excretion in Ctl mice, whereas these changes were suppressed but urinary Ang II was increased in conditional MegKO mice. CONCLUSION Increased glomerular filtration of AGT is likely to augment Ang II production in the proximal tubular lumen. Thus, megalin-dependent Ang II uptake should be involved in the ERK1/2 signaling that activates proximal tubular NHE3 in vivo , thereby causing Na + retention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Ryohei Kaseda
- Department of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata
| | | | | | - Ichiei Narita
- Department of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata
| | - Kouhei Tsumoto
- The Institute of Medical Science and Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
31
|
Shastri S, Patel J, Sambandam KK, Lederer ED. Kidney Stone Pathophysiology, Evaluation and Management: Core Curriculum 2023. Am J Kidney Dis 2023; 82:617-634. [PMID: 37565942 PMCID: PMC11370273 DOI: 10.1053/j.ajkd.2023.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/31/2023] [Indexed: 08/12/2023]
Abstract
Kidney stone disease, also known as nephrolithiasis or urolithiasis, is a disorder in which urinary solutes precipitate to form aggregates of crystalline material in the urinary space. The incidence of nephrolithiasis has been increasing, and the demographics have been evolving. Once viewed as a limited disease with intermittent exacerbations that are simply managed by urologists, nephrolithiasis is now recognized as a complex condition requiring thorough evaluation and multifaceted care. Kidney stones are frequently manifestations of underlying systemic medical conditions such as the metabolic syndrome, genetic disorders, or endocrinopathies. Analysis of urine chemistries and stone composition provide a window into pathogenesis and direct ancillary studies to uncover underlying diseases. These studies allow providers to devise individualized strategies to limit future stone events. Given its complexity, kidney stone disease is best addressed by a team led by nephrologists and urologists with input from multiple other health professionals including dietitians, endocrinologists, interventional radiologists, and endocrine surgeons. In this installment of AJKD's Core Curriculum in Nephrology, we provide a case-based overview of nephrolithiasis, divided by the individual stone types. The reader will gain a pragmatic understanding of the pathophysiology, evaluation, and management of this condition.
Collapse
Affiliation(s)
| | - Jiten Patel
- UT Southwestern Medical Center, Dallas, Texas
| | | | - Eleanor D Lederer
- UT Southwestern Medical Center, Dallas, Texas; VA North Texas Health Care Services, Dallas, Texas.
| |
Collapse
|
32
|
Ferreira RM, de Almeida R, Culp C, Witzmann F, Wang M, Kher R, Nagami GT, Mohallem R, Andolino CJ, Aryal UK, Eadon MT, Bacallao RL. Proteomic analysis of murine kidney proximal tubule sub-segment derived cell lines reveals preferences in mitochondrial pathway activity. J Proteomics 2023; 289:104998. [PMID: 37657718 PMCID: PMC10843797 DOI: 10.1016/j.jprot.2023.104998] [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: 04/19/2023] [Revised: 07/16/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
The proximal tubule (PT) is a nephron segment that is responsible for the majority of solute and water reabsorption in the kidney. Each of its sub-segments have specialized functions; however, little is known about the genes and proteins that determine the oxidative phosphorylation capacity of the PT sub-segments. This information is critical to understanding kidney function and will provide a comprehensive landscape of renal cell adaptations to injury, physiologic stressors, and development. This study analyzed three immortalized murine renal cell lines (PT S1, S2, and S3 segments) for protein content and compared them to a murine fibroblast cell line. All three proximal tubule cell lines generate ATP predominantly by oxidative phosphorylation while the fibroblast cell line is glycolytic. The proteomic data demonstrates that the most significant difference in proteomic signatures between the cell lines are proteins known to be localized in the nucleus followed by mitochondrial proteins. Mitochondrial metabolic substrate utilization assays were performed using the proximal tubule cell lines to determine substrate utilization kinetics thereby providing a physiologic context to the proteomic dataset. This data will allow researchers to study differences in nephron-specific cell lines, between epithelial and fibroblast cells, and between actively respiring cells and glycolytic cells. SIGNIFICANCE: Proteomic analysis of proteins expressed in immortalized murine renal proximal tubule cells was compared to a murine fibroblast cell line proteome. The proximal tubule segment specific cell lines: S1, S2 and S3 are all grown under conditions whereby the cells generate ATP by oxidative phosphorylation while the fibroblast cell line utilizes anaerobic glycolysis for ATP generation. The proteomic studies allow for the following queries: 1) comparisons between the proximal tubule segment specific cell lines, 2) comparisons between polarized epithelia and fibroblasts, 3) comparison between cells employing oxidative phosphorylation versus anaerobic glycolysis and 4) comparisons between cells grown on clear versus opaque membrane supports. The data finds major differences in nuclear protein expression and mitochondrial proteins. This proteomic data set will be an important baseline dataset for investigators who need immortalized renal proximal tubule epithelial cells for their research.
Collapse
Affiliation(s)
- Ricardo Melo Ferreira
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Rita de Almeida
- Instituto de Física and Instituto Nacional de Ciência e Tecnologia, Universidade Federal do Rio Grande do Sul, 91501-970 Porto Alegre, RS, Brazil.
| | - Clayton Culp
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Frank Witzmann
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mu Wang
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Rajesh Kher
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Glenn T Nagami
- Division of Nephrology, VA Greater Los Angeles Healthcare System, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| | - Rodrigo Mohallem
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA; Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.
| | - Chaylen Jade Andolino
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.
| | - Uma K Aryal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA; Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.
| | - Michael T Eadon
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Robert L Bacallao
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
33
|
Hsiao HY, Yen TH, Wu FY, Cheng CM, Liu JW, Fan YT, Huang JJ, Nien CY. Delivery and Transcriptome Assessment of an In Vitro Three-Dimensional Proximal Tubule Model Established by Human Kidney 2 Cells in Clinical Gelatin Sponges. Int J Mol Sci 2023; 24:15547. [PMID: 37958530 PMCID: PMC10650118 DOI: 10.3390/ijms242115547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 11/15/2023] Open
Abstract
The high prevalence of kidney diseases and the low identification rate of drug nephrotoxicity in preclinical studies reinforce the need for representative yet feasible renal models. Although in vitro cell-based models utilizing renal proximal tubules are widely used for kidney research, many proximal tubule cell (PTC) lines have been indicated to be less sensitive to nephrotoxins, mainly due to altered expression of transporters under a two-dimensional culture (2D) environment. Here, we selected HK-2 cells to establish a simplified three-dimensional (3D) model using gelatin sponges as scaffolds. In addition to cell viability and morphology, we conducted a comprehensive transcriptome comparison and correlation analysis of 2D and 3D cultured HK-2 cells to native human PTCs. Our 3D model displayed stable and long-term growth with a tubule-like morphology and demonstrated a more comparable gene expression profile to native human PTCs compared to the 2D model. Many missing or low expressions of major genes involved in PTC transport and metabolic processes were restored, which is crucial for successful nephrotoxicity prediction. Consequently, we established a cost-effective yet more representative model for in vivo PTC studies and presented a comprehensive transcriptome analysis for the systematic characterization of PTC lines.
Collapse
Affiliation(s)
- Hui-Yi Hsiao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Tzung-Hai Yen
- Department of Nephrology, Clinical Poison Center, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Department of Nephrology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Fang-Yu Wu
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300193, Taiwan;
| | - Jia-Wei Liu
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Yu-Ting Fan
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Jung-Ju Huang
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Chung-Yi Nien
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| |
Collapse
|
34
|
Lindner ML, Lohmeyer JL, Adam EH, Zacharowski K, Weber CF. [Mechanisms of action and clinical application of diuretics in intensive care medicine]. DIE ANAESTHESIOLOGIE 2023; 72:757-770. [PMID: 37768358 DOI: 10.1007/s00101-023-01338-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 09/29/2023]
Abstract
The paired kidneys play a significant role in the human body due to the multitude of physiological tasks. Complex biochemical processes keep the sensitive electrolyte and water balance stable and thus ensure the organism's ability to adapt to exogenous and endogenous factors, which is essential for survival. The drug class of diuretics includes substances with very differing pharmacological characteristics. The functioning of the nephron is therefore indispensable for a deeper understanding of the pharmacodynamics, pharmacokinetics and side effect profile of diuretics. In the treatment of acute heart failure with pulmonary congestion, certain diuretics represent an important therapeutic option to counteract hypervolemia and thus an increase in preload. According to current data, diuretics have no proven benefits in the treatment or prevention of acute kidney injury but they can counteract hypervolemia and under certain conditions even reduce the use of renal replacement procedures.
Collapse
Affiliation(s)
- M-L Lindner
- Abteilung für Anästhesiologie, Intensiv- und Notfallmedizin, Asklepios Kliniken Hamburg GmbH, Asklepios Klinik Wandsbek, Alphonsstraße 14, 22043, Hamburg, Deutschland.
| | - J L Lohmeyer
- Abteilung für Anästhesiologie, Intensiv‑, Notfall-, und Schmerzmedizin, Asklepios Kliniken Hamburg GmbH, Asklepios Klinik Altona, Paul-Ehrlich-Straße 1, 22763, Hamburg, Deutschland
| | - E H Adam
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Frankfurt, Goethe-Universität, Theodor Stern Kai 7, 60590, Frankfurt, Deutschland
| | - K Zacharowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Frankfurt, Goethe-Universität, Theodor Stern Kai 7, 60590, Frankfurt, Deutschland
| | - C F Weber
- Abteilung für Anästhesiologie, Intensiv- und Notfallmedizin, Asklepios Kliniken Hamburg GmbH, Asklepios Klinik Wandsbek, Alphonsstraße 14, 22043, Hamburg, Deutschland
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Frankfurt, Goethe-Universität, Theodor Stern Kai 7, 60590, Frankfurt, Deutschland
| |
Collapse
|
35
|
Sahu A, Min K, Jeon SH, Kwon K, Tae G. Self-assembled hemin-conjugated heparin with dual-enzymatic cascade reaction activities for acute kidney injury. Carbohydr Polym 2023; 316:121088. [PMID: 37321716 DOI: 10.1016/j.carbpol.2023.121088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/16/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023]
Abstract
Nanozymes have prominent catalytic activities with high stability as a substitute for unstable and expensive natural enzymes. However, most nanozymes are metal/inorganic nanomaterials, facing difficulty in clinical translation due to their unproven biosafety and limited biodegradability issues. Hemin, an organometallic porphyrin, was newly found to possess superoxide dismutase (SOD) mimetic activity along with previously known catalase (CAT) mimetic activity. However, hemin has poor bioavailability due to its low water solubility. Therefore, a highly biocompatible and biodegradable organic-based nanozyme system with SOD/CAT mimetic cascade reaction activity was developed by conjugating hemin to heparin (HepH) or chitosan (CS-H). Between them, Hep-H formed a smaller (<50 nm) and more stable self-assembled nanostructure and even possessed much higher and more stable SOD and CAT activities as well as the cascade reaction activity compared to CS-H and free hemin. Hep-H also showed a better cell protection effect against reactive oxygen species (ROS) compared to CS-H and hemin in vitro. Furthermore, Hep-H was selectively delivered to the injured kidney upon intravenous administration at the analysis time point (24 h) and exhibited excellent therapeutic effects on an acute kidney injury model by efficiently removing ROS, reducing inflammation, and minimizing structural and functional damage to the kidney.
Collapse
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kiyoon Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
36
|
Khalil NN, Petersen AP, Song CJ, Chen Y, Takamoto K, Kellogg AC, Chen EZ, McMahon AP, McCain ML. User-friendly microfluidic system reveals native-like morphological and transcriptomic phenotypes induced by shear stress in proximal tubule epithelium. APL Bioeng 2023; 7:036106. [PMID: 37584027 PMCID: PMC10424157 DOI: 10.1063/5.0143614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
Drug-induced nephrotoxicity is a leading cause of drug attrition, partly due to the limited relevance of pre-clinical models of the proximal tubule. Culturing proximal tubule epithelial cells (PTECs) under fluid flow to mimic physiological shear stress has been shown to improve select phenotypes, but existing flow systems are expensive and difficult to implement by non-experts in microfluidics. Here, we designed and fabricated an accessible and modular flow system for culturing PTECs under physiological shear stress, which induced native-like cuboidal morphology, downregulated pathways associated with hypoxia, stress, and injury, and upregulated xenobiotic metabolism pathways. We also compared the expression profiles of shear-dependent genes in our in vitro PTEC tissues to that of ex vivo proximal tubules and observed stronger clustering between ex vivo proximal tubules and PTECs under physiological shear stress relative to PTECs under negligible shear stress. Together, these data illustrate the utility of our user-friendly flow system and highlight the role of shear stress in promoting native-like morphological and transcriptomic phenotypes in PTECs in vitro, which is critical for developing more relevant pre-clinical models of the proximal tubule for drug screening or disease modeling.
Collapse
Affiliation(s)
- Natalie N. Khalil
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Andrew P. Petersen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Yibu Chen
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, California 90089, USA
| | - Kaelyn Takamoto
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Austin C. Kellogg
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Elaine Zhelan Chen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Megan L. McCain
- Author to whom correspondence should be addressed:. Tel.: +1 2138210791. URL:https://livingsystemsengineering.usc.edu
| |
Collapse
|
37
|
Keuper-Navis M, Walles M, Poller B, Myszczyszyn A, van der Made TK, Donkers J, Eslami Amirabadi H, Wilmer MJ, Aan S, Spee B, Masereeuw R, van de Steeg E. The application of organ-on-chip models for the prediction of human pharmacokinetic profiles during drug development. Pharmacol Res 2023; 195:106853. [PMID: 37473876 DOI: 10.1016/j.phrs.2023.106853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Organ-on-chip (OoC) technology has led to in vitro models with many new possibilities compared to conventional in vitro and in vivo models. In this review, the potential of OoC models to improve the prediction of human oral bioavailability and intrinsic clearance is discussed, with a focus on the functionality of the models and the application in current drug development practice. Multi-OoC models demonstrating the application for pharmacokinetic (PK) studies are summarized and existing challenges are identified. Physiological parameters for a minimal viable platform of a multi-OoC model to study PK are provided, together with PK specific read-outs and recommendations for relevant reference compounds to validate the model. Finally, the translation to in vivo PK profiles is discussed, which will be required to routinely apply OoC models during drug development.
Collapse
Affiliation(s)
- Marit Keuper-Navis
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Markus Walles
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Birk Poller
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Adam Myszczyszyn
- Faculty of Veterinary Medicine & Regenerative Medicine Center Utrecht (RMCU), Utrecht University, Utrecht, the Netherlands
| | - Thomas K van der Made
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Joanne Donkers
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | | | | | - Saskia Aan
- Stichting Proefdiervrij, Den Haag, the Netherlands
| | - Bart Spee
- Faculty of Veterinary Medicine & Regenerative Medicine Center Utrecht (RMCU), Utrecht University, Utrecht, the Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands.
| |
Collapse
|
38
|
Hao Y, Zhao L, Zhao JY, Han X, Zhou X. Unveiling the potential of mitochondrial dynamics as a therapeutic strategy for acute kidney injury. Front Cell Dev Biol 2023; 11:1244313. [PMID: 37635869 PMCID: PMC10456901 DOI: 10.3389/fcell.2023.1244313] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Acute Kidney Injury (AKI), a critical clinical syndrome, has been strongly linked to mitochondrial malfunction. Mitochondria, vital cellular organelles, play a key role in regulating cellular energy metabolism and ensuring cell survival. Impaired mitochondrial function in AKI leads to decreased energy generation, elevated oxidative stress, and the initiation of inflammatory cascades, resulting in renal tissue damage and functional impairment. Therefore, mitochondria have gained significant research attention as a potential therapeutic target for AKI. Mitochondrial dynamics, which encompass the adaptive shifts of mitochondria within cellular environments, exert significant influence on mitochondrial function. Modulating these dynamics, such as promoting mitochondrial fusion and inhibiting mitochondrial division, offers opportunities to mitigate renal injury in AKI. Consequently, elucidating the mechanisms underlying mitochondrial dynamics has gained considerable importance, providing valuable insights into mitochondrial regulation and facilitating the development of innovative therapeutic approaches for AKI. This comprehensive review aims to highlight the latest advancements in mitochondrial dynamics research, provide an exhaustive analysis of existing studies investigating the relationship between mitochondrial dynamics and acute injury, and shed light on their implications for AKI. The ultimate goal is to advance the development of more effective therapeutic interventions for managing AKI.
Collapse
Affiliation(s)
- Yajie Hao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Limei Zhao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Jing Yu Zhao
- The Third Clinical College, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, China
| | - Xiutao Han
- The Third Clinical College, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, China
| | - Xiaoshuang Zhou
- Department of Nephrology, Shanxi Provincial People’s Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Shanxi Kidney Disease Institute, Taiyuan, China
| |
Collapse
|
39
|
Radi SH, Vemuri K, Martinez-Lomeli J, Sladek FM. HNF4α isoforms: the fraternal twin master regulators of liver function. Front Endocrinol (Lausanne) 2023; 14:1226173. [PMID: 37600688 PMCID: PMC10438950 DOI: 10.3389/fendo.2023.1226173] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
In the more than 30 years since the purification and cloning of Hepatocyte Nuclear Factor 4 (HNF4α), considerable insight into its role in liver function has been gleaned from its target genes and mouse experiments. HNF4α plays a key role in lipid and glucose metabolism and intersects with not just diabetes and circadian rhythms but also with liver cancer, although much remains to be elucidated about those interactions. Similarly, while we are beginning to elucidate the role of the isoforms expressed from its two promoters, we know little about the alternatively spliced variants in other portions of the protein and their impact on the 1000-plus HNF4α target genes. This review will address how HNF4α came to be called the master regulator of liver-specific gene expression with a focus on its role in basic metabolism, the contributions of the various isoforms and the intriguing intersection with the circadian clock.
Collapse
Affiliation(s)
- Sarah H. Radi
- Department of Biochemistry, University of California, Riverside, Riverside, CA, United States
| | - Kiranmayi Vemuri
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Jose Martinez-Lomeli
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Frances M. Sladek
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
40
|
Novella-Rausell C, Grudniewska M, Peters DJ, Mahfouz A. A comprehensive mouse kidney atlas enables rare cell population characterization and robust marker discovery. iScience 2023; 26:106877. [PMID: 37275529 PMCID: PMC10238935 DOI: 10.1016/j.isci.2023.106877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/24/2023] [Accepted: 05/10/2023] [Indexed: 06/07/2023] Open
Abstract
The kidney's cellular diversity is on par with its physiological intricacy; yet identifying cell populations and their markers remains challenging. Here, we created a comprehensive atlas of the healthy adult mouse kidney (MKA: Mouse Kidney Atlas) by integrating 140.000 cells and nuclei from 59 publicly available single-cell and single-nuclei RNA-sequencing datasets from eight independent studies. To harmonize annotations across datasets, we built a hierarchical model of the cell populations. Our model allows the incorporation of novel cell populations and the refinement of known profiles as more datasets become available. Using MKA and the learned model of cellular hierarchies, we predicted previously missing cell annotations from several studies. The MKA allowed us to identify reproducible markers across studies for poorly understood cell types and transitional states, which we verified using existing data from micro-dissected samples and spatial transcriptomics.
Collapse
Affiliation(s)
- Claudio Novella-Rausell
- Department of Human Genetics, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
- GenomeScan, 2333 BZ Leiden, the Netherlands
| | | | - Dorien J.M. Peters
- Department of Human Genetics, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Ahmed Mahfouz
- Department of Human Genetics, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
41
|
Verissimo T, Dalga D, Arnoux G, Sakhi I, Faivre A, Auwerx H, Bourgeois S, Paolucci D, Gex Q, Rutkowski JM, Legouis D, Wagner CA, Hall AM, de Seigneux S. PCK1 is a key regulator of metabolic and mitochondrial functions in renal tubular cells. Am J Physiol Renal Physiol 2023; 324:F532-F543. [PMID: 37102687 PMCID: PMC10202477 DOI: 10.1152/ajprenal.00038.2023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/28/2023] Open
Abstract
Phosphoenolpyruvate carboxykinase 1 (PCK1 or PEPCK-C) is a cytosolic enzyme converting oxaloacetate to phosphoenolpyruvate, with a potential role in gluconeogenesis, ammoniagenesis, and cataplerosis in the liver. Kidney proximal tubule cells display high expression of this enzyme, whose importance is currently not well defined. We generated PCK1 kidney-specific knockout and knockin mice under the tubular cell-specific PAX8 promoter. We studied the effect of PCK1 deletion and overexpression at the renal level on tubular physiology under normal conditions and during metabolic acidosis and proteinuric renal disease. PCK1 deletion led to hyperchloremic metabolic acidosis characterized by reduced but not abolished ammoniagenesis. PCK1 deletion also resulted in glycosuria, lactaturia, and altered systemic glucose and lactate metabolism at baseline and during metabolic acidosis. Metabolic acidosis resulted in kidney injury in PCK1-deficient animals with decreased creatinine clearance and albuminuria. PCK1 further regulated energy production by the proximal tubule, and PCK1 deletion decreased ATP generation. In proteinuric chronic kidney disease, mitigation of PCK1 downregulation led to better renal function preservation. PCK1 is essential for kidney tubular cell acid-base control, mitochondrial function, and glucose/lactate homeostasis. Loss of PCK1 increases tubular injury during acidosis. Mitigating kidney tubular PCK1 downregulation during proteinuric renal disease improves renal function.NEW & NOTEWORTHY Phosphoenolpyruvate carboxykinase 1 (PCK1) is highly expressed in the proximal tubule. We show here that this enzyme is crucial for the maintenance of normal tubular physiology, lactate, and glucose homeostasis. PCK1 is a regulator of acid-base balance and ammoniagenesis. Preventing PCK1 downregulation during renal injury improves renal function, rendering it an important target during renal disease.
Collapse
Affiliation(s)
- Thomas Verissimo
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| | - Delal Dalga
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| | - Grégoire Arnoux
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Imene Sakhi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Anna Faivre
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Hannah Auwerx
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Deborah Paolucci
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Quentin Gex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | | | - David Legouis
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Division of Intensive Care, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Sophie de Seigneux
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
42
|
Koh ES. Hidden Acid Retention with Normal Serum Bicarbonate Level in Chronic Kidney Disease. Electrolyte Blood Press 2023; 21:34-43. [PMID: 37434806 PMCID: PMC10329907 DOI: 10.5049/ebp.2023.21.1.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/06/2023] [Accepted: 04/16/2023] [Indexed: 07/13/2023] Open
Abstract
Management of metabolic acidosis is crucial for preserving bone, muscle, and renal health, as evidenced by the results of several interventional studies conducted on patients with chronic kidney disease (CKD). Considering the continuity of CKD progression over time, it is reasonable to deduce that a subclinical form of metabolic acidosis may exist prior to the manifestation of overt metabolic acidosis. Covert H+ retention with normal serum bicarbonate level in patients with CKD may result in maladaptive responses that contribute to kidney function deterioration, even in the early stages of the disease. The loss of adaptive compensatory mechanisms of urinary acid excretion may be a key factor in this process. Early modulation of these responses could be an important therapeutic strategy in preventing CKD progression. However, to date, the optimal approach for alkali therapy in subclinical metabolic acidosis in CKD remains uncertain. There is a lack of established guidelines on when to initiate alkali therapy, potential side effects of alkali agents, and the optimal blood bicarbonate levels based on evidence-based practices. Therefore, further research is necessary to address these concerns and establish more robust guidelines for the use of alkali therapy in patients with CKD. Herein, we provide an overview of recent developments on this subject and examine the potential therapeutic approaches that interventional treatments may present for patients with hidden H+ retention, exhibiting normal serum bicarbonate levels - commonly described as subclinical or eubicarbonatemic metabolic acidosis in patients with CKD.
Collapse
Affiliation(s)
- Eun Sil Koh
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
43
|
Banan Sadeghian R, Ueno R, Takata Y, Kawakami A, Ma C, Araoka T, Takasato M, Yokokawa R. Cells sorted off hiPSC-derived kidney organoids coupled with immortalized cells reliably model the proximal tubule. Commun Biol 2023; 6:483. [PMID: 37142732 PMCID: PMC10160057 DOI: 10.1038/s42003-023-04862-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Of late, numerous microphysiological systems have been employed to model the renal proximal tubule. Yet there is lack of research on refining the functions of the proximal tubule epithelial layer-selective filtration and reabsorption. In this report, pseudo proximal tubule cells extracted from human-induced pluripotent stem cell-derived kidney organoids are combined and cultured with immortalized proximal tubule cells. It is shown that the cocultured tissue is an impervious epithelium that offers improved levels of certain transporters, extracellular matrix proteins collagen and laminin, and superior glucose transport and P-glycoprotein activity. mRNA expression levels higher than those obtained from each cell type were detected, suggesting an anomalous synergistic crosstalk between the two. Alongside, the improvements in morphological characteristics and performance of the immortalized proximal tubule tissue layer exposed, upon maturation, to human umbilical vein endothelial cells are thoroughly quantified and compared. Glucose and albumin reabsorption, as well as xenobiotic efflux rates through P-glycoprotein were all improved. The data presented abreast highlight the advantages of the cocultured epithelial layer and the non-iPSC-based bilayer. The in vitro models presented herein can be helpful in personalized nephrotoxicity studies.
Collapse
Affiliation(s)
| | - Ryohei Ueno
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Yuji Takata
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Akihiko Kawakami
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Cheng Ma
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Minoru Takasato
- RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, 650-0047, Japan
- Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan.
| |
Collapse
|
44
|
Tang H, Xu C, Zhang P, Luo T, Huang Y, Yang X. A profile of SGLT-2 inhibitors in hyponatremia: The evidence to date. Eur J Pharm Sci 2023; 184:106415. [PMID: 36870579 DOI: 10.1016/j.ejps.2023.106415] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/14/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Hyponatremia is the most common electrolyte disorder in clinical practice, which may lead to life-threatening complications. Several lines of evidence suggest that hyponatremia is associated not only with significant increases in length of stay, cost, and financial burden, but also with increased morbidity and mortality. Hyponatremia is also considered to be a negative prognostic factor in patients with heart failure and cancer. Although multiple therapeutic methods are available for treating hyponatremia, most have some limitations, such as poor compliance, rapid correction of serum Na+, other negative side effects and high cost. Given these limitations, identifying novel therapies for hyponatremia is essential. Recent clinical studies have shown that SGLT-2 inhibitors (SGLT 2i) significantly increased serum Na+ levels and were well tolerated by patients who underwent this treatment. Therefore, oral administration of SGLT 2i appears to be an effective treatment for hyponatremia. This article will briefly review the etiology of hyponatremia and integrated control of sodium within the kidney, current therapies for hyponatremia, potential mechanisms and efficacy of SGLT 2i for hyponatremia, and the benefits in cardiovascular, cancer, and kidney disease by regulating sodium and water balance.
Collapse
Affiliation(s)
- Hui Tang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Changjing Xu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Piao Zhang
- Department of Pharmacy, Ya 'an People's Hospital, Ya 'an, Sichuan 646000, China
| | - Taimin Luo
- Department of pharmacy, Chengdu Seventh People's Hospital, Chengdu, Sichuan 610000, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
45
|
Zhou W, Simic P, Zhou IY, Caravan P, Vela Parada X, Wen D, Washington OL, Shvedova M, Pierce KA, Clish CB, Mannstadt M, Kobayashi T, Wein MN, Jüppner H, Rhee EP. Kidney glycolysis serves as a mammalian phosphate sensor that maintains phosphate homeostasis. J Clin Invest 2023; 133:e164610. [PMID: 36821389 PMCID: PMC10104895 DOI: 10.1172/jci164610] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 02/21/2023] [Indexed: 02/24/2023] Open
Abstract
How phosphate levels are detected in mammals is unknown. The bone-derived hormone fibroblast growth factor 23 (FGF23) lowers blood phosphate levels by reducing kidney phosphate reabsorption and 1,25(OH)2D production, but phosphate does not directly stimulate bone FGF23 expression. Using PET scanning and LC-MS, we found that phosphate increases kidney-specific glycolysis and synthesis of glycerol-3-phosphate (G-3-P), which then circulates to bone to trigger FGF23 production. Further, we found that G-3-P dehydrogenase 1 (Gpd1), a cytosolic enzyme that synthesizes G-3-P and oxidizes NADH to NAD+, is required for phosphate-stimulated G-3-P and FGF23 production and prevention of hyperphosphatemia. In proximal tubule cells, we found that phosphate availability is substrate-limiting for glycolysis and G-3-P production and that increased glycolysis and Gpd1 activity are coupled through cytosolic NAD+ recycling. Finally, we show that the type II sodium-dependent phosphate cotransporter Npt2a, which is primarily expressed in the proximal tubule, conferred kidney specificity to phosphate-stimulated G-3-P production. Importantly, exogenous G-3-P stimulated FGF23 production when Npt2a or Gpd1 were absent, confirming that it was the key circulating factor downstream of glycolytic phosphate sensing in the kidney. Together, these findings place glycolysis at the nexus of mineral and energy metabolism and identify a kidney-bone feedback loop that controls phosphate homeostasis.
Collapse
Affiliation(s)
- Wen Zhou
- Nephrology Division, Department of Medicine, and
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Petra Simic
- Nephrology Division, Department of Medicine, and
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Iris Y. Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Xavier Vela Parada
- Nephrology Division, Department of Medicine, and
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Donghai Wen
- Nephrology Division, Department of Medicine, and
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Onica L. Washington
- Nephrology Division, Department of Medicine, and
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Maria Shvedova
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kerry A. Pierce
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Michael Mannstadt
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tatsuya Kobayashi
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marc N. Wein
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Harald Jüppner
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Pediatric Nephrology Unit, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eugene P. Rhee
- Nephrology Division, Department of Medicine, and
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Hsu CN, Hsuan CF, Liao D, Chang JKJ, Chang AJW, Hee SW, Lee HL, Teng SIF. Anti-Diabetic Therapy and Heart Failure: Recent Advances in Clinical Evidence and Molecular Mechanism. Life (Basel) 2023; 13:1024. [PMID: 37109553 PMCID: PMC10144651 DOI: 10.3390/life13041024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Diabetic patients have a two- to four-fold increase in the risk of heart failure (HF), and the co-existence of diabetes and HF is associated with poor prognosis. In randomized clinical trials (RCTs), compelling evidence has demonstrated the beneficial effects of sodium-glucose co-transporter-2 inhibitors on HF. The mechanism includes increased glucosuria, restored tubular glomerular feedback with attenuated renin-angiotensin II-aldosterone activation, improved energy utilization, decreased sympathetic tone, improved mitochondria calcium homeostasis, enhanced autophagy, and reduced cardiac inflammation, oxidative stress, and fibrosis. The RCTs demonstrated a neutral effect of the glucagon-like peptide receptor agonist on HF despite its weight-reducing effect, probably due to it possibly increasing the heart rate via increasing cyclic adenosine monophosphate (cAMP). Observational studies supported the markedly beneficial effects of bariatric and metabolic surgery on HF despite no current supporting evidence from RCTs. Bromocriptine can be used to treat peripartum cardiomyopathy by reducing the harmful cleaved prolactin fragments during late pregnancy. Preclinical studies suggest the possible beneficial effect of imeglimin on HF through improving mitochondrial function, but further clinical evidence is needed. Although abundant preclinical and observational studies support the beneficial effects of metformin on HF, there is limited evidence from RCTs. Thiazolidinediones increase the risk of hospitalized HF through increasing renal tubular sodium reabsorption mediated via both the genomic and non-genomic action of PPARγ. RCTs suggest that dipeptidyl peptidase-4 inhibitors, including saxagliptin and possibly alogliptin, may increase the risk of hospitalized HF, probably owing to increased circulating vasoactive peptides, which impair endothelial function, activate sympathetic tones, and cause cardiac remodeling. Observational studies and RCTs have demonstrated the neutral effects of insulin, sulfonylureas, an alpha-glucosidase inhibitor, and lifestyle interventions on HF in diabetic patients.
Collapse
Affiliation(s)
- Chih-Neng Hsu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin 640, Taiwan
| | - Chin-Feng Hsuan
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan
- Division of Cardiology, Department of Internal Medicine, E-Da Dachang Hospital, I-Shou University, Kaohsiung 824, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 840, Taiwan
| | - Daniel Liao
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Jack Keng-Jui Chang
- Biological Programs for Younger Scholar, Academia Sinica, Taipei 115, Taiwan
| | - Allen Jiun-Wei Chang
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Siow-Wey Hee
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Hsiao-Lin Lee
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Sean I. F. Teng
- Department of Cardiology, Ming-Sheng General Hospital, Taoyuan 330, Taiwan
| |
Collapse
|
47
|
Huyan Y, Wang C, Kang H, Chen X, Chang Y, Liu S, Song J. Single-Cell Transcriptome Sequencing Reveals Molecular Mechanisms of Renal Injury in Essential Hypertension. Kidney Blood Press Res 2023; 48:297-313. [PMID: 37062270 PMCID: PMC10308540 DOI: 10.1159/000530624] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
INTRODUCTION Hypertensive nephropathy is characterized by glomerular and tubulointerstitial damage, but we know little about changes in cell-specific gene expression in the early stages of hypertensive kidney injury, which usually has no obvious pathological changes. METHODS We performed unbiased single-cell RNA sequencing of rat kidney samples from hypertensive kidney injury to generate 10,602 single-cell transcriptomes from 2 control and 2 early stage hypertensive kidney injury samples. RESULTS All major cell types of the kidney were represented in the final dataset. Side-by-side comparisons showed that cell type-specific changes in gene expression are critical for functional impairment of glomeruli and tubules and activation of immune cells. In particular, we found a significantly reduced gene expression profile of maintaining vascular integrity in glomerular cells of hypertensive kidney injury. Meanwhile, the expression of genes associated with oxidative stress injury and fibrosis in the renal tubules and collecting ducts was elevated, but the degree of tubular cells response to injury differed between parts. We also found a signature of immune cell infiltration in hypertensive kidney injury. CONCLUSION Exploring the changes of gene expression in hypertension-injured kidneys may be helpful to identify the early biomarkers and signal pathways of this disease. Our data provide rich resources for understanding the pathogenesis of hypertensive renal injury and formulating effective treatment strategies.
Collapse
Affiliation(s)
- Yige Huyan
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunyue Wang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongen Kang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Chang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sheng Liu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
48
|
Shinya S, Muthusinghe DS, Koizumi N, Yoshimatsu K, Nakayama SMM, Ishizuka M, Ikenaka Y. Effects of Diazinon Exposure on Urinary Shedding of Leptospira interrogans Serogroup Hebdomadis in Mice. TOXICS 2023; 11:361. [PMID: 37112589 PMCID: PMC10141796 DOI: 10.3390/toxics11040361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/14/2023] [Accepted: 04/03/2023] [Indexed: 06/19/2023]
Abstract
Wild rodents are natural hosts of Leptospira spp. and are exposed to various pesticides, some of which are immunotoxic. Rodent urine is an important source of infection for humans and other animals. We evaluated the effects of pesticide exposure on Leptospira growth in mice. Diazinon, at doses of 0.2, 1, and 5 mg/kg/day, was orally administered continuously to mice infected with Leptospira interrogans serogroup Hebdomadis for 32 days. The numbers of L. interrogans in urine and kidney tissues were significantly lower in mice exposed to 5 mg/kg/day diazinon than in unexposed mice (p < 0.05). The urinary concentration of 2-isopropyl-6-methyl-4-pyrimidinol, the metabolite of diazinon, was comparable with the concentration at which viability of L. interrogans was decreased in in vitro assay, suggesting that it had toxic effects on L. interrogans in the proximal renal tubules. Diazinon exposure reinforced Leptospira-induced expression of inflammatory cytokine genes in kidney tissues, and an enhanced immune system might suppress Leptospira growth. These results suggest that diazinon exposure may not increase the risk of Leptospira transmission to humans through mice. This novel study evaluated the relationship between pesticide exposure and Leptospira infection in mice, and the results could be useful for risk assessment of leptospirosis.
Collapse
Affiliation(s)
- So Shinya
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | | | - Nobuo Koizumi
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan
| | - Kumiko Yoshimatsu
- Institute of Genetic Medicine, Hokkaido University, Sapporo 060-0808, Japan
| | - Shouta M. M. Nakayama
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Biomedical Science Department, School of Veterinary Medicine, The University of Zambia, Lusaka 32379, Zambia
| | - Mayumi Ishizuka
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan
- One Health Research Center, Hokkaido University, M18, W9, Kita-ku, Sapporo 060-0818, Japan
- Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom 2520, South Africa
| |
Collapse
|
49
|
Wagner CA, Unwin R, Lopez-Garcia SC, Kleta R, Bockenhauer D, Walsh S. The pathophysiology of distal renal tubular acidosis. Nat Rev Nephrol 2023; 19:384-400. [PMID: 37016093 DOI: 10.1038/s41581-023-00699-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
The kidneys have a central role in the control of acid-base homeostasis owing to bicarbonate reabsorption and production of ammonia and ammonium in the proximal tubule and active acid secretion along the collecting duct. Impaired acid excretion by the collecting duct system causes distal renal tubular acidosis (dRTA), which is characterized by the failure to acidify urine below pH 5.5. This defect originates from reduced function of acid-secretory type A intercalated cells. Inherited forms of dRTA are caused by variants in SLC4A1, ATP6V1B1, ATP6V0A4, FOXI1, WDR72 and probably in other genes that are yet to be discovered. Inheritance of dRTA follows autosomal-dominant and -recessive patterns. Acquired forms of dRTA are caused by various types of autoimmune diseases or adverse effects of some drugs. Incomplete dRTA is frequently found in patients with and without kidney stone disease. These patients fail to appropriately acidify their urine when challenged, suggesting that incomplete dRTA may represent an intermediate state in the spectrum of the ability to excrete acids. Unrecognized or insufficiently treated dRTA can cause rickets and failure to thrive in children, osteomalacia in adults, nephrolithiasis and nephrocalcinosis. Electrolyte disorders are also often present and poorly controlled dRTA can increase the risk of developing chronic kidney disease.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK.
| | - Robert Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Sergio C Lopez-Garcia
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Robert Kleta
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Detlef Bockenhauer
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Stephen Walsh
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| |
Collapse
|
50
|
Piret SE, Mallipattu SK. Transcriptional regulation of proximal tubular metabolism in acute kidney injury. Pediatr Nephrol 2023; 38:975-986. [PMID: 36181578 DOI: 10.1007/s00467-022-05748-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/07/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022]
Abstract
The kidney, and in particular the proximal tubule (PT), has a high demand for ATP, due to its function in bulk reabsorption of solutes. In normal PT, ATP levels are predominantly maintained by fatty acid β-oxidation (FAO), the tricarboxylic acid (TCA) cycle, and oxidative phosphorylation. The normal PT also undertakes gluconeogenesis and metabolism of amino acids. Acute kidney injury (AKI) results in profound PT metabolic alterations, including suppression of FAO, gluconeogenesis, and metabolism of some amino acids, and upregulation of glycolytic enzymes. Recent studies have elucidated new transcriptional mechanisms regulating metabolic pathways in normal PT, as well as the metabolic switch in AKI. A number of transcription factors have been shown to play important roles in FAO, which are themselves downregulated in AKI, while hypoxia-inducible factor 1α, which is upregulated in ischemia-reperfusion injury, is a likely driver of the upregulation of glycolytic enzymes. Transcriptional regulation of amino acid metabolic pathways is less well understood, except for catabolism of branched-chain amino acids, which is likely suppressed in AKI by upregulation of Krüppel-like factor 6. This review will focus on the transcriptional regulation of specific metabolic pathways in normal PT and in AKI, as well as highlighting some of the gaps in knowledge and challenges that remain to be addressed.
Collapse
Affiliation(s)
- Sian E Piret
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, 101 Nicolls Road, Stony Brook, NY, 11794, USA.
| | - Sandeep K Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, 101 Nicolls Road, Stony Brook, NY, 11794, USA
- Renal Division, Northport VA Medical Center, Northport, NY, USA
| |
Collapse
|