1
|
Rossano AJ, Zhang L, Anderson JB, Holmes HL, Mandal AK, Decker JW, Mount DB, Romero MF. Ex vivo quantification of intracellular pH in Drosophila Malpighian tubule reveals basolateral HCO 3 -/oxalate exchange through a novel oxalate transporter "Neat". Front Physiol 2025; 16:1468451. [PMID: 40356774 PMCID: PMC12066474 DOI: 10.3389/fphys.2025.1468451] [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: 07/22/2024] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Nephrolithiasis is a painful and costly healthcare complication. The most common kidney stones are composed of calcium oxalate and thus renal handling of oxalate is an important facet of understanding the pathogenesis of nephrolithiasis. Recently, the Drosophila melanogaster Malpighian tubule (MT) has emerged as a robust model of trans-epithelial ion transport and nephrolithiasis as MTs readily form luminal calcium-oxalate crystals in the presence of oxalate. Drosophila Prestin (dPrestin, Slc26a6) transports oxalate across the apical surface of the MT into the lumen but a full model of the trans-epithelial movement of oxalate (Ox2-) in the Drosophila MT has been lacking as the basolateral oxalate transporter has remained uncharacterized. Methods The objective of this work was to identify and characterize the Drosophila basolateral Ox2- transporter through ex vivo real-time quantification of intracellular pH (pHi) and Xenopus oocyte transport assays. Results A putative basolateral oxalate transporter CG5002 ("Neat") was identified through sequence homology and displayed robust Cl--independent Ox2- transport and electroneutral Ox2- transport in Xenopus oocytes. pHi in extracted fly MTs was monitored by using the GAL4/UAS system to selectively express pHerry, a pseudo-ratiometric genetically-encoded pH indicator (GEpHI) in the cytosol of the principal cells of the MT. Basolateral perfusion of MTs in CO2/HCO3 --buffered solution produced a large acidification followed by rapid recovery in the transitional segment of the anterior MT. Recovery was interrupted by basolateral application of 1 mM Ox2- or 1 mM SO4 2. Tissue specific knock-down of Neat with interference RNA (RNAi) reduced the rate of acid-loading in the transitional segment of the MT with regard to Ox2- and SO4 2-. Knockdown of Neat in the MT also significantly reduced luminal calcium oxalate crystal formation in a fly ex vivo model of calcium oxalate nephrolithiasis. Discussion These data indicate Neat is a significant Drosophila basolateral MT oxalate transporter and the basolateral movement of oxalate is functionally coupled to movement of acid equivalents, potentially as Ox2-/HCO3 - exchange, Ox2-/OH- exchange, or Ox2-:H+ co-transport.
Collapse
Affiliation(s)
- Adam J. Rossano
- Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Lili Zhang
- Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Jacob B. Anderson
- Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Heather L. Holmes
- Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Asim K. Mandal
- Renal Division, Brigham and Womens Hospital and Harvard Medical School, Boston, MA, United States
| | - James W. Decker
- Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Biomedical Engineering, Boston University, Boston, MA, United States
| | - David B. Mount
- Renal Division, Brigham and Womens Hospital and Harvard Medical School, Boston, MA, United States
- Renal Division, West Roxbury VA Medical Center, West Roxbury, MA, United States
| | - Michael F. Romero
- Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
- Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| |
Collapse
|
2
|
Krogsaeter EK, McKetney J, Valiente-Banuet L, Marquez A, Willis A, Cakir Z, Stevenson E, Jang GM, Rao A, Li E, Zhou A, Attili A, Chang TS, Kampmann M, Huang Y, Krogan NJ, Swaney DL. Lysosomal proteomics reveals mechanisms of neuronal apoE4-associated lysosomal dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.02.560519. [PMID: 37873080 PMCID: PMC10592882 DOI: 10.1101/2023.10.02.560519] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
ApoE4 is the primary risk factor for Alzheimer Disease (AD). Early AD pathological events first affect the neuronal endolysosomal system, which in turn causes neuronal protein aggregation and cell death. Despite the crucial influence of lysosomes upon AD pathophysiology, and that apoE4 localizes to lysosomes, the influence of apoE4 on lysosomal function remains unexplored. We find that expression of apoE4 in neuronal cell lines results in lysosomal alkalinization and impaired lysosomal function. To identify driving factors for these defects, we performed quantitative lysosomal proteome profiling. This revealed that apoE4 expression results in differential regulation of numerous lysosomal proteins, correlating with apoE allele status and disease severity in AD brains. In particular, apoE4 expression results in the depletion of lysosomal Lgals3bp and the accumulation of lysosomal Tmed5. We additionally validated that these lysosomal protein changes can be targeted to modulate lysosomal function. Taken together, this work thereby reveals that apoE4 causes widespread lysosomal defects through remodeling the lysosomal proteome, with the lysosomal Tmed5 accumulation and Lgals3bp depletion manifesting as lysosomal alkalinization in apoE4 neurons.
Collapse
Affiliation(s)
- Einar K. Krogsaeter
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- These authors contributed equally
| | - Justin McKetney
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- These authors contributed equally
| | - Leopoldo Valiente-Banuet
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Angelica Marquez
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Alexandra Willis
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Zeynep Cakir
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Erica Stevenson
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Gwendolyn M. Jang
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Antara Rao
- Gladstone Institute of Neurological Disease, The J. David Gladstone Institutes, San Francisco, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, USA
| | - Emmy Li
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, USA
| | - Anton Zhou
- Gladstone Institute of Neurological Disease, The J. David Gladstone Institutes, San Francisco, USA
| | - Anjani Attili
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California, USA
- Biosciences Internship Program, City College of San Francisco, USA
| | - Timothy S. Chang
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, The J. David Gladstone Institutes, San Francisco, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, USA
- Neuroscience Graduate Program, University of California, San Francisco, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, USA
- Departments of Neurology and Pathology, University of California, San Francisco, USA
| | - Nevan J. Krogan
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Danielle L. Swaney
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| |
Collapse
|
3
|
Zhang L, Leonard N, Passaro R, Luan MS, Van Tuyen P, Han LTN, Cam NH, Vogelnest L, Lynch M, Fine AE, Nga NTT, Van Long N, Rawson BM, Behie A, Van Nguyen T, Le MD, Nadler T, Walter L, Marques-Bonet T, Hofreiter M, Li M, Liu Z, Roos C. Genomic adaptation to small population size and saltwater consumption in the critically endangered Cat Ba langur. Nat Commun 2024; 15:8531. [PMID: 39358348 PMCID: PMC11447269 DOI: 10.1038/s41467-024-52811-7] [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: 04/26/2023] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Many mammal species have declining populations, but the consequences of small population size on the genomic makeup of species remain largely unknown. We investigated the evolutionary history, genetic load and adaptive potential of the Cat Ba langur (Trachypithecus poliocephalus), a primate species endemic to Vietnam's famous Ha Long Bay and with less than 100 living individuals one of the most threatened primates in the world. Using high-coverage whole genome data of four wild individuals, we revealed the Cat Ba langur as sister species to its conspecifics of the northern limestone langur clade and found no evidence for extensive secondary gene flow after their initial separation. Compared to other primates and mammals, the Cat Ba langur showed low levels of genetic diversity, long runs of homozygosity, high levels of inbreeding and an excess of deleterious mutations in homozygous state. On the other hand, genetic diversity has been maintained in protein-coding genes and on the gene-rich human chromosome 19 ortholog, suggesting that the Cat Ba langur retained most of its adaptive potential. The Cat Ba langur also exhibits several unique non-synonymous variants that are related to calcium and sodium metabolism, which may have improved adaptation to high calcium intake and saltwater consumption.
Collapse
Affiliation(s)
- Liye Zhang
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany.
- International Max Planck Research School for Genome Science (IMPRS-GS), University of Göttingen, Göttingen, Germany.
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Neahga Leonard
- Cat Ba Langur Conservation Project (CBLCP), Cat Ba National Park, Cat Ba Island, Cat Hai District, Hai Phong Province, Vietnam
| | - Rick Passaro
- Cat Ba Langur Conservation Project (CBLCP), Cat Ba National Park, Cat Ba Island, Cat Hai District, Hai Phong Province, Vietnam
| | - Mai Sy Luan
- Cat Ba Langur Conservation Project (CBLCP), Cat Ba National Park, Cat Ba Island, Cat Hai District, Hai Phong Province, Vietnam
| | - Pham Van Tuyen
- Cat Ba Langur Conservation Project (CBLCP), Cat Ba National Park, Cat Ba Island, Cat Hai District, Hai Phong Province, Vietnam
| | - Le Thi Ngoc Han
- Cat Ba Langur Conservation Project (CBLCP), Cat Ba National Park, Cat Ba Island, Cat Hai District, Hai Phong Province, Vietnam
| | - Nguyen Huy Cam
- Cat Ba Langur Conservation Project (CBLCP), Cat Ba National Park, Cat Ba Island, Cat Hai District, Hai Phong Province, Vietnam
| | - Larry Vogelnest
- Taronga Conservation Society Australia, Mosman, NSW, Australia
| | - Michael Lynch
- Melbourne Zoo, Zoos Victoria, Parkville, VIC, Australia
| | - Amanda E Fine
- Wildlife Conservation Society (WCS), Health Program, New York, NY, USA
| | | | - Nguyen Van Long
- Wildlife Conservation Society (WCS), Vietnam Country Program, Hanoi, Vietnam
| | - Benjamin M Rawson
- World Wildlife Fund for Nature (WWF) International, Gland, Switzerland
| | - Alison Behie
- School of Archaeology and Anthropology, The Australian National University, Canberra, ACT, Australia
| | - Truong Van Nguyen
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- Evolutionary Adaptive Genomics, Institute of Biochemistry and Biology, Department of Science, University of Potsdam, Potsdam, Germany
- Central Institute for Natural Resources and Environmental Studies, Vietnam National University, Hanoi, Vietnam
| | - Minh D Le
- Central Institute for Natural Resources and Environmental Studies, Vietnam National University, Hanoi, Vietnam
- Faculty of Environmental Sciences, University of Science, Vietnam National University, Hanoi, Vietnam
| | - Tilo Nadler
- Three Monkeys Wildlife Conservancy, Nho Quan District, Ninh Binh Province, Ninh Binh, Vietnam
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Tomas Marques-Bonet
- Institute of Evolutionary Biology (UPF-CSIC), PRBB, Barcelona, Spain
- Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Spain
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, Edifici ICTA-ICP, Cerdanyola del Vallès, Spain
| | - Michael Hofreiter
- Evolutionary Adaptive Genomics, Institute of Biochemistry and Biology, Department of Science, University of Potsdam, Potsdam, Germany.
| | - Ming Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Zhijin Liu
- College of Life Sciences, Capital Normal University, Beijing, China.
| | - Christian Roos
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany.
- Gene Bank of Primates, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany.
| |
Collapse
|
4
|
Raut S, Singh K, Sanghvi S, Loyo-Celis V, Varghese L, Singh E, Gururaja Rao S, Singh H. Chloride ions in health and disease. Biosci Rep 2024; 44:BSR20240029. [PMID: 38573803 PMCID: PMC11065649 DOI: 10.1042/bsr20240029] [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: 01/09/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/06/2024] Open
Abstract
Chloride is a key anion involved in cellular physiology by regulating its homeostasis and rheostatic processes. Changes in cellular Cl- concentration result in differential regulation of cellular functions such as transcription and translation, post-translation modifications, cell cycle and proliferation, cell volume, and pH levels. In intracellular compartments, Cl- modulates the function of lysosomes, mitochondria, endosomes, phagosomes, the nucleus, and the endoplasmic reticulum. In extracellular fluid (ECF), Cl- is present in blood/plasma and interstitial fluid compartments. A reduction in Cl- levels in ECF can result in cell volume contraction. Cl- is the key physiological anion and is a principal compensatory ion for the movement of the major cations such as Na+, K+, and Ca2+. Over the past 25 years, we have increased our understanding of cellular signaling mediated by Cl-, which has helped in understanding the molecular and metabolic changes observed in pathologies with altered Cl- levels. Here, we review the concentration of Cl- in various organs and cellular compartments, ion channels responsible for its transportation, and recent information on its physiological roles.
Collapse
Affiliation(s)
- Satish K. Raut
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Kulwinder Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Shridhar Sanghvi
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
- Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, U.S.A
| | - Veronica Loyo-Celis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Liyah Varghese
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | - Ekam R. Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
| | | | - Harpreet Singh
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, U.S.A
- Department of Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, U.S.A
| |
Collapse
|
5
|
Lee D, Hong JH. Chloride/Multiple Anion Exchanger SLC26A Family: Systemic Roles of SLC26A4 in Various Organs. Int J Mol Sci 2024; 25:4190. [PMID: 38673775 PMCID: PMC11050216 DOI: 10.3390/ijms25084190] [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: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Solute carrier family 26 member 4 (SLC26A4) is a member of the SLC26A transporter family and is expressed in various tissues, including the airway epithelium, kidney, thyroid, and tumors. It transports various ions, including bicarbonate, chloride, iodine, and oxalate. As a multiple-ion transporter, SLC26A4 is involved in the maintenance of hearing function, renal function, blood pressure, and hormone and pH regulation. In this review, we have summarized the various functions of SLC26A4 in multiple tissues and organs. Moreover, the relationships between SLC26A4 and other channels, such as cystic fibrosis transmembrane conductance regulator, epithelial sodium channel, and sodium chloride cotransporter, are highlighted. Although the modulation of SLC26A4 is critical for recovery from malfunctions of various organs, development of specific inducers or agonists of SLC26A4 remains challenging. This review contributes to providing a better understanding of the role of SLC26A4 and development of therapeutic approaches for the SLC26A4-associated hearing loss and SLC26A4-related dysfunction of various organs.
Collapse
Affiliation(s)
| | - Jeong Hee Hong
- Department of Health Sciences and Technology, GAIHST (Gachon Advanced Institute for Health Sciences and Technology), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| |
Collapse
|
6
|
Geertsma ER, Oliver D. SLC26 Anion Transporters. Handb Exp Pharmacol 2024; 283:319-360. [PMID: 37947907 DOI: 10.1007/164_2023_698] [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: 11/12/2023]
Abstract
Solute carrier family 26 (SLC26) is a family of functionally diverse anion transporters found in all kingdoms of life. Anions transported by SLC26 proteins include chloride, bicarbonate, and sulfate, but also small organic dicarboxylates such as fumarate and oxalate. The human genome encodes ten functional homologs, several of which are causally associated with severe human diseases, highlighting their physiological importance. Here, we review novel insights into the structure and function of SLC26 proteins and summarize the physiological relevance of human members.
Collapse
Affiliation(s)
- Eric R Geertsma
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, Marburg, Giessen, Germany.
| |
Collapse
|
7
|
Wei S, Chen B, Low SW, Poore CP, Gao Y, Nilius B, Liao P. SLC26A11 Inhibition Reduces Oncotic Neuronal Death and Attenuates Stroke Reperfusion Injury. Mol Neurobiol 2023; 60:5931-5943. [PMID: 37380823 PMCID: PMC10471688 DOI: 10.1007/s12035-023-03453-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Neuronal swelling is a pathological feature of stroke which contributes to the formation of cytotoxic edema. Under hypoxic condition, aberrant accumulation of sodium and chloride ions inside neurons increases osmotic pressure, leading to cell volume increase. Sodium entry pathway in neurons has been studied extensively. Here, we determine whether SLC26A11 is the major chloride entry pathway under hypoxia and could be the target for protection against ischemic stroke. In this study, electrophysiological properties of chloride current in primary cultured neurons were characterized using low chloride solution, 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid, and SLC26A11-specific siRNA under physiological conditions or ATP-depleted conditions. In vivo effect of SLC26A11 was evaluated on a rat stroke reperfusion model. We found that SLC26A11 mRNA in primary cultured neurons was upregulated as early as 6 h after oxygen glucose deprivation, and later, the protein level was elevated accordingly. Blockade of SLC26A11 activity could reduce chloride entry and attenuate hypoxia-induced neuronal swelling. In the animal stroke model, SLC26A11 upregulation was mainly located in surviving neurons close to the infarct core. SLC26A11 inhibition ameliorates infarct formation and improves functional recovery. These findings demonstrate that SLC26A11 is a major pathway for chloride entry in stroke, contributing to neuronal swelling. Inhibition of SLC26A11 could be a novel therapeutic strategy for stroke.
Collapse
Affiliation(s)
- Shunhui Wei
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Bo Chen
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - See Wee Low
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Charlene Priscilla Poore
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
| | - Yahui Gao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Present Address: Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 119077 Singapore
| | - Bernd Nilius
- Department of Cellular and Molecular Medicine, KU Leuven, 3000 Louvain, Belgium
| | - Ping Liao
- Calcium Signalling Laboratory, Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433 Singapore
- Duke-NUS Medical School, Singapore, 169857 Singapore
- Health and Social Sciences, Singapore Institute of Technology, Singapore, 138683 Singapore
| |
Collapse
|
8
|
Chelangarimiyandoab F, Mungara P, Batta M, Cordat E. Urinary Tract Infections: Renal Intercalated Cells Protect against Pathogens. J Am Soc Nephrol 2023; 34:1605-1614. [PMID: 37401780 PMCID: PMC10561816 DOI: 10.1681/asn.0000000000000187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
Urinary tract infections affect more than 1 in 2 women during their lifetime. Among these, more than 10% of patients carry antibiotic-resistant bacterial strains, highlighting the urgent need to identify alternative treatments. While innate defense mechanisms are well-characterized in the lower urinary tract, it is becoming evident that the collecting duct (CD), the first renal segment encountered by invading uropathogenic bacteria, also contributes to bacterial clearance. However, the role of this segment is beginning to be understood. This review summarizes the current knowledge on CD intercalated cells in urinary tract bacterial clearance. Understanding the innate protective role of the uroepithelium and of the CD offers new opportunities for alternative therapeutic strategies.
Collapse
Affiliation(s)
- Forough Chelangarimiyandoab
- Department of Physiology and Membrane Protein Disease Research Group, Faculty of Medicine & Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
9
|
Ahmed SBM, Radwan N, Amer S, Saheb Sharif-Askari N, Mahdami A, Samara KA, Halwani R, Jelinek HF. Assessing the Link between Diabetic Metabolic Dysregulation and Breast Cancer Progression. Int J Mol Sci 2023; 24:11816. [PMID: 37511575 PMCID: PMC10380477 DOI: 10.3390/ijms241411816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetes mellitus is a burdensome disease that affects various cellular functions through altered glucose metabolism. Several reports have linked diabetes to cancer development; however, the exact molecular mechanism of how diabetes-related traits contribute to cancer progression is not fully understood. The current study aimed to explore the molecular mechanism underlying the potential effect of hyperglycemia combined with hyperinsulinemia on the progression of breast cancer cells. To this end, gene dysregulation induced by the exposure of MCF7 breast cancer cells to hyperglycemia (HG), or a combination of hyperglycemia and hyperinsulinemia (HGI), was analyzed using a microarray gene expression assay. Hyperglycemia combined with hyperinsulinemia induced differential expression of 45 genes (greater than or equal to two-fold), which were not shared by other treatments. On the other hand, in silico analysis performed using a publicly available dataset (GEO: GSE150586) revealed differential upregulation of 15 genes in the breast tumor tissues of diabetic patients with breast cancer when compared with breast cancer patients with no diabetes. SLC26A11, ALDH1A3, MED20, PABPC4 and SCP2 were among the top upregulated genes in both microarray data and the in silico analysis. In conclusion, hyperglycemia combined with hyperinsulinemia caused a likely unique signature that contributes to acquiring more carcinogenic traits. Indeed, these findings might potentially add emphasis on how monitoring diabetes-related metabolic alteration as an adjunct to diabetes therapy is important in improving breast cancer outcomes. However, further detailed studies are required to decipher the role of the highlighted genes, in this study, in the pathogenesis of breast cancer in patients with a different glycemic index.
Collapse
Affiliation(s)
- Samrein B M Ahmed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Health, Wellbeing and Life Sciences, Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield S1 1WB, UK
| | - Nada Radwan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sara Amer
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amena Mahdami
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Kamel A Samara
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rabih Halwani
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Herbert F Jelinek
- Department of Biomedical Engineering and Health Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
10
|
Mégier C, Dumery G, Luton D. Iodine and Thyroid Maternal and Fetal Metabolism during Pregnancy. Metabolites 2023; 13:metabo13050633. [PMID: 37233673 DOI: 10.3390/metabo13050633] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Thyroid hormones and iodine are required to increase basal metabolic rate and to regulate protein synthesis, long bone growth and neuronal maturation. They are also essential for protein, fat and carbohydrate metabolism regulation. Imbalances in thyroid and iodine metabolism can negatively affect these vital functions. Pregnant women are at risk of hypo or hyperthyroidism, in relation to or regardless of their medical history, with potential dramatic outcomes. Fetal development highly relies on thyroid and iodine metabolism and can be compromised if they malfunction. As the interface between the fetus and the mother, the placenta plays a crucial role in thyroid and iodine metabolism during pregnancy. This narrative review aims to provide an update on current knowledge of thyroid and iodine metabolism in normal and pathological pregnancies. After a brief description of general thyroid and iodine metabolism, their main modifications during normal pregnancies and the placental molecular actors are described. We then discuss the most frequent pathologies to illustrate the upmost importance of iodine and thyroid for both the mother and the fetus.
Collapse
Affiliation(s)
- Charles Mégier
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Grégoire Dumery
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Dominique Luton
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| |
Collapse
|
11
|
Barone S, Brooks M, Zahedi K, Holliday LS, Bissler J, Yu JJ, Soleimani M. Identification of an Electrogenic 2Cl -/H + Exchanger, ClC5, as a Chloride-Secreting Transporter Candidate in Kidney Cyst Epithelium in Tuberous Sclerosis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:191-200. [PMID: 36336066 PMCID: PMC9926528 DOI: 10.1016/j.ajpath.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/23/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Kidney cyst expansion in tuberous sclerosis complex (TSC) or polycystic kidney disease (PKD) requires active secretion of chloride (Cl-) into the cyst lumen. In PKD, Cl- secretion is primarily mediated via the cystic fibrosis transmembrane conductance regulator (CFTR) in principal cells. Kidney cystogenesis in TSC is predominantly composed of type A intercalated cells, which do not exhibit noticeable expression of CFTR. The identity of the Cl--secreting molecule(s) in TSC cyst epithelia remains speculative. RNA-sequencing analysis results were used to examine the expression of FOXi1, the chief regulator of acid base transporters in intercalated cells, along with localization of Cl- channel 5 (ClC5), in various models of TSC. Results from Tsc2+/- mice showed that the expansion of kidney cysts corresponded to the induction of Foxi1 and correlated with the appearance of ClC5 and H+-ATPase on the apical membrane of cyst epithelia. In various mouse models of TSC, Foxi1 was robustly induced in the kidney, and ClC5 and H+-ATPase were expressed on the apical membrane of cyst epithelia. Expression of ClC5 was also detected on the apical membrane of cyst epithelia in humans with TSC but was absent in humans with autosomal dominant PKD or in a mouse model of PKD. These results indicate that ClC5 is expressed on the apical membrane of cyst epithelia and is a likely candidate mediating Cl- secretion into the kidney cyst lumen in TSC.
Collapse
Affiliation(s)
- Sharon Barone
- Research Services, Veterans Health Care Medical Center, Albuquerque, New Mexico; Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Marybeth Brooks
- Research Services, Veterans Health Care Medical Center, Albuquerque, New Mexico; Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Kamyar Zahedi
- Research Services, Veterans Health Care Medical Center, Albuquerque, New Mexico; Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | | | - John Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, Tennessee; Department of Pediatrics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jane J Yu
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Manoocher Soleimani
- Research Services, Veterans Health Care Medical Center, Albuquerque, New Mexico; Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.
| |
Collapse
|
12
|
Xu J, Barone S, Varasteh Kia M, Holliday LS, Zahedi K, Soleimani M. Identification of IQGAP1 as a SLC26A4 (Pendrin)-Binding Protein in the Kidney. Front Mol Biosci 2022; 9:874186. [PMID: 35601831 PMCID: PMC9117723 DOI: 10.3389/fmolb.2022.874186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Several members of the SLC26A family of transporters, including SLC26A3 (DRA), SLC26A5 (prestin), SLC26A6 (PAT-1; CFEX) and SLC26A9, form multi-protein complexes with a number of molecules (e.g., cytoskeletal proteins, anchoring or adaptor proteins, cystic fibrosis transmembrane conductance regulator, and protein kinases). These interactions provide regulatory signals for these molecules. However, the identity of proteins that interact with the Cl-/HCO3 - exchanger, SLC26A4 (pendrin), have yet to be determined. The purpose of this study is to identify the protein(s) that interact with pendrin. Methods: A yeast two hybrid (Y2H) system was employed to screen a mouse kidney cDNA library using the C-terminal fragment of SLC26A4 as bait. Immunofluorescence microscopic examination of kidney sections, as well as co-immunoprecipitation assays, were performed using affinity purified antibodies and kidney protein extracts to confirm the co-localization and interaction of pendrin and the identified binding partners. Co-expression studies were carried out in cultured cells to examine the effect of binding partners on pendrin trafficking and activity. Results: The Y2H studies identified IQ motif-containing GTPase-activating protein 1 (IQGAP1) as a protein that binds to SLC26A4's C-terminus. Co-immunoprecipitation experiments using affinity purified anti-IQGAP1 antibodies followed by western blot analysis of kidney protein eluates using pendrin-specific antibodies confirmed the interaction of pendrin and IQGAP1. Immunofluorescence microscopy studies demonstrated that IQGAP1 co-localizes with pendrin on the apical membrane of B-intercalated cells, whereas it shows basolateral expression in A-intercalated cells in the cortical collecting duct (CCD). Functional and confocal studies in HEK-293 cells, as well as confocal studies in MDCK cells, demonstrated that the co-transfection of pendrin and IQGAP1 shows strong co-localization of the two molecules on the plasma membrane along with enhanced Cl-/HCO3 - exchanger activity. Conclusion: IQGAP1 was identified as a protein that binds to the C-terminus of pendrin in B-intercalated cells. IQGAP1 co-localized with pendrin on the apical membrane of B-intercalated cells. Co-expression of IQGAP1 with pendrin resulted in strong co-localization of the two molecules and increased the activity of pendrin in the plasma membrane in cultured cells. We propose that pendrin's interaction with IQGAP1 may play a critical role in the regulation of CCD function and physiology, and that disruption of this interaction could contribute to altered pendrin trafficking and/or activity in pathophysiologic states.
Collapse
Affiliation(s)
- Jie Xu
- Research Services, VA Medical Center, Albuquerque, NM, United States,Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Sharon Barone
- Research Services, VA Medical Center, Albuquerque, NM, United States,Department of Medicine, University of Cincinnati, Cincinnati, OH, United States,Department of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Mujan Varasteh Kia
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - L. Shannon Holliday
- Department of Orthodontics, University of Florida, Gainesville, FL, United States
| | - Kamyar Zahedi
- Research Services, VA Medical Center, Albuquerque, NM, United States,Department of Medicine, University of Cincinnati, Cincinnati, OH, United States,Department of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Manoocher Soleimani
- Research Services, VA Medical Center, Albuquerque, NM, United States,Department of Medicine, University of Cincinnati, Cincinnati, OH, United States,Department of Medicine, University of New Mexico, Albuquerque, NM, United States,*Correspondence: Manoocher Soleimani,
| |
Collapse
|
13
|
Zhu J, Mou Y, Ye S, Hu H, Wang R, Yang Q, Hu Y. Identification of a Six-Gene SLC Family Signature With Prognostic Value in Patients With Lung Adenocarcinoma. Front Cell Dev Biol 2022; 9:803198. [PMID: 34977043 PMCID: PMC8714960 DOI: 10.3389/fcell.2021.803198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022] Open
Abstract
Given the importance of solute carrier (SLC) proteins in maintaining cellular metabolic homeostasis and that their dysregulation contributes to cancer progression, here we constructed a robust SLC family signature for lung adenocarcinoma (LUAD) patient stratification. Transcriptomic profiles and relevant clinical information of LUAD patients were downloaded from the TCGA and GEO databases. SLC family genes differentially expressed between LUAD tissues and adjacent normal tissues were identified using limma in R. Of these, prognosis-related SLC family genes were further screened out and used to construct a novel SLC family-based signature in the training cohort. The accuracy of the prognostic signature was assessed in the testing cohort, the entire cohort, and the external GSE72094 cohort. Correlations between the prognostic signature and the tumor immune microenvironment and immune cell infiltrates were further explored. We found that seventy percent of SLC family genes (279/397) were differentially expressed between LUAC tissues and adjacent normal. Twenty-six genes with p-values < 0.05 in univariate Cox regression analysis and Kaplan-Meier survival analysis were regarded as prognosis-related SLC family genes, six of which were used to construct a prognostic signature for patient classification into high- and low-risk groups. Kaplan-Meier survival analysis in all internal and external cohorts revealed a better overall survival for patients in the low-risk group than those in the high-risk group. Univariate and multivariate Cox regression analyses indicated that the derived risk score was an independent prognostic factor for LUAD patients. Moreover, a nomogram based on the six-gene signature and clinicopathological factors was developed for clinical application. High-risk patients had lower stromal, immune, and ESTIMATE scores and higher tumor purities than those in the low-risk group. The proportions of infiltrating naive CD4 T cells, activated memory CD4 T cells, M0 macrophages, resting dendritic cells, resting mast cells, activated mast cells, and eosinophils were significantly different between the high- and low-risk prognostic groups. In all, the six-gene SLC family signature is of satisfactory accuracy and generalizability for predicting overall survival in patients with LUAD. Furthermore, this prognostics signature is related to tumor immune status and distinct immune cell infiltrates in the tumor microenvironment.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Mou
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenglan Ye
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongling Hu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rujuan Wang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Yang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Hu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Insights into the Interaction of Lysosomal Amino Acid Transporters SLC38A9 and SLC36A1 Involved in mTORC1 Signaling in C2C12 Cells. Biomolecules 2021; 11:biom11091314. [PMID: 34572527 PMCID: PMC8467208 DOI: 10.3390/biom11091314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022] Open
Abstract
Amino acids are critical for mammalian target of rapamycin complex 1 (mTORC1) activation on the lysosomal surface. Amino acid transporters SLC38A9 and SLC36A1 are the members of the lysosomal amino acid sensing machinery that activates mTORC1. The current study aims to clarify the interaction of SLC38A9 and SLC36A1. Here, we discovered that leucine increased expressions of SLC38A9 and SLC36A1, leading to mTORC1 activation. SLC38A9 interacted with SLC36A1 and they enhanced each other's expression levels and locations on the lysosomal surface. Additionally, the interacting proteins of SLC38A9 in C2C12 cells were identified to participate in amino acid sensing mechanism, mTORC1 signaling pathway, and protein synthesis, which provided a resource for future investigations of skeletal muscle mass.
Collapse
|
15
|
Kidney intercalated cells and the transcription factor FOXi1 drive cystogenesis in tuberous sclerosis complex. Proc Natl Acad Sci U S A 2021; 118:2020190118. [PMID: 33536341 PMCID: PMC8017711 DOI: 10.1073/pnas.2020190118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by mutations in TSC1 or TSC2 gene and affects multiple organs, including the kidney, where it presents with angiomyolipomata and cysts that can result in kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Current studies demonstrate that kidney cyst epithelia in TSC mouse models and in humans with TSC are composed of hyperproliferating intercalated cells, along with activation of H+-ATPase and carbonic anhydrase 2. Interfering with intercalated cell proliferation completely inhibited and inactivating carbonic anhydrase 2 significantly protected against cyst formation in TSC. Targeting the acid base and/or electrolyte transporters of intercalated cells may provide a therapeutic approach for the treatment of kidney cysts in TSC. Tuberous sclerosis complex (TSC) is caused by mutations in either TSC1 or TSC2 genes and affects multiple organs, including kidney, lung, and brain. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomata) and cysts, which eventually leads to kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Here, we report that mice with principal cell-specific inactivation of Tsc1 develop numerous cortical cysts, which are overwhelmingly composed of hyperproliferating A-intercalated (A-IC) cells. RNA sequencing and confirmatory expression studies demonstrated robust expression of Forkhead Transcription Factor 1 (Foxi1) and its downstream targets, apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in cyst epithelia in Tsc1 knockout (KO) mice but not in Pkd1 mutant mice. In addition, the electrogenic 2Cl−/H+ exchanger (CLC-5) is significantly up-regulated and shows remarkable colocalization with H+-ATPase on the apical membrane of cyst epithelia in Tsc1 KO mice. Deletion of Foxi1, which is vital to intercalated cells viability and H+-ATPase expression, completely abrogated the cyst burden in Tsc1 KO mice, as indicated by MRI images and histological analysis in kidneys of Foxi1/Tsc1 double-knockout (dKO) mice. Deletion of CAII, which is critical to H+-ATPase activation, caused significant reduction in cyst burden and increased life expectancy in CAII/Tsc1 dKO mice vs. Tsc1 KO mice. We propose that intercalated cells and their acid/base/electrolyte transport machinery (H+-ATPase/CAII/CLC-5) are critical to cystogenesis, and their inhibition or inactivation is associated with significant protection against cyst generation and/or enlargement in TSC.
Collapse
|
16
|
Yu Q. Slc26a3 (DRA) in the Gut: Expression, Function, Regulation, Role in Infectious Diarrhea and Inflammatory Bowel Disease. Inflamm Bowel Dis 2021; 27:575-584. [PMID: 32989468 DOI: 10.1093/ibd/izaa256] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The transport of transepithelial Cl- and HCO3- is crucial for the function of the intestinal epithelium and maintains the acid-based homeostasis. Slc26a3 (DRA), as a key chloride-bicarbonate exchanger protein in the intestinal epithelial luminal membrane, participates in the electroneutral NaCl absorption of intestine, together with Na+/H+ exchangers. Increasing recent evidence supports the essential role of decreased DRA function or expression in infectious diarrhea and inflammatory bowel disease (IBD). METHOD In this review, we give an overview of the current knowledge of Slc26a3, including its cloning and expression, function, roles in infectious diarrhea and IBD, and mechanisms of actions. A better understanding of the physiological and pathophysiological relevance of Slc26a3 in infectious diarrhea and IBD may reveal novel targets for future therapy. CONCLUSION Understanding the physiological function, regulatory interactions, and the potential mechanisms of Slc26a3 in the pathophysiology of infectious diarrhea and IBD will define novel therapeutic approaches in future.
Collapse
Affiliation(s)
- Qin Yu
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan City, China
| |
Collapse
|
17
|
Zhang F, Zhong W, Li H, Huang K, Yu M, Liu Y. TP53 Mutational Status-Based Genomic Signature for Prognosis and Predicting Therapeutic Response in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:665265. [PMID: 34124046 PMCID: PMC8187932 DOI: 10.3389/fcell.2021.665265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/09/2021] [Indexed: 02/05/2023] Open
Abstract
TP53 mutation is a critical driver mutation that affects the carcinogenesis and prognosis of patients with pancreatic cancer (PC). Currently, there is no driver mutation-derived signature based on TP53 mutational status for prognosis and predicting therapeutic response in PC. In the present study, we characterized the TP53 mutational phenotypes in multiple patient cohorts and developed a prognostic TP53-associated signature based on differentially expressed genes between PC samples with mutated TP53 and wild-type TP53. Comprehensive investigations were carried out in prognostic stratification, genetic variation, immune cell infiltration, and efficacy prediction of chemotherapy and targeted therapy. We found that TP53 mutation commonly occurred as a survival-related driver mutation in PC. In total, 1,154 differentially expressed genes were found between two distinct TP53 mutational phenotypes. A five-gene TP53-associated signature was constructed in The Cancer Genome Atlas (TCGA) cohort by least absolute shrinkage and selection operator (LASSO)-Cox analysis and proven to be a robust prognostic predictor, which performed well in three independent Gene Expression Omnibus (GEO) validating cohorts. Remarkably, patients in the low-risk group were characterized with decreased tumor mutation burden and activity of immunity, with favorable prognosis. Higher fractions of macrophages M0 and impaired CD8 + T cells were observed in patients in the high-risk group, suggesting immunosuppression with poor survival. Patients in the high-risk group also demonstrated enhanced response to specific chemotherapeutic agents, including gemcitabine and paclitaxel. Several targeted inhibitors, like histamine receptor inhibitor, were screened out as promising drugs for PC treatment. Collectively, the TP53-associated signature is a novel prognostic biomarker and predictive indicator of PC. The signature could contribute to optimizing prognostic stratification and guide effective PC treatments.
Collapse
Affiliation(s)
- Feng Zhang
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Wenhui Zhong
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Honghao Li
- The Sixth Affiliated Hospital, Sun Yat-sen University Guangdong Gastrointestinal Hospital, Guangzhou, China
| | - Kaijun Huang
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Min Yu
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Min Yu,
| | - Yubin Liu
- Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Yubin Liu,
| |
Collapse
|
18
|
Astapenko D, Navratil P, Pouska J, Cerny V. Clinical physiology aspects of chloremia in fluid therapy: a systematic review. Perioper Med (Lond) 2020; 9:40. [PMID: 33298166 PMCID: PMC7727154 DOI: 10.1186/s13741-020-00171-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/30/2020] [Indexed: 11/10/2022] Open
Abstract
Background This systematic review discusses a clinical physiology aspect of chloride in fluid therapy. Crystalloid solutions are one of the most widely used remedies. While generally used in medicine for almost 190 years, studies focused largely on their safety have only been published since the new millennium. The most widely used solution, normal saline, is most often referred to in this context. Its excessive administration results in hyperchloremic metabolic acidosis with other consequences, including higher mortality rates. Methods Original papers and review articles eligible for developing the present paper were identified by searching online in the electronic MEDLINE database. The keywords searched for included hyperchloremia, hypochloremia, and compound words containing the word “chloride,” infusion therapy, metabolic acidosis, renal failure, and review. Results A total of 21,758 papers published before 31 May 2020 were identified; of this number, 630 duplicates were removed from the list. Upon excluding articles based on their title or abstract, 1850 papers were screened, of which 63 full-text articles were assessed. Conclusions According to the latest medical concepts, dyschloremia (both hyperchloremia and hypochloremia) represents a factor indisputably having a negative effect on selected variables of clinical outcome. As infusion therapy can significantly impact chloride homeostasis of the body, the choice of infusion solutions should always take into account the potentially adverse impact of chloride content on chloremia and organ function.
Collapse
Affiliation(s)
- David Astapenko
- Department of Anesthesiology, Resuscitation and Intensive Care Medicine, University Hospital Hradec Kralove, Sokolská 581, 500 05, Hradec Kralove, Czech Republic. .,Faculty of Medicine in Hradec Kralove, Charles University, Prague, Czech Republic.
| | - Pavel Navratil
- Faculty of Medicine in Hradec Kralove, Charles University, Prague, Czech Republic.,Department of Urology, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jiri Pouska
- Department of Anesthesiology, Resuscitation and Intensive Care Medicine, University Hospital Plzen, Plzen, Czech Republic.,Faculty of Medicine in Plzen, Charles University, Plzen, Czech Republic
| | - Vladimir Cerny
- Department of Anesthesiology, Resuscitation and Intensive Care Medicine, University Hospital Hradec Kralove, Sokolská 581, 500 05, Hradec Kralove, Czech Republic.,Faculty of Medicine in Hradec Kralove, Charles University, Prague, Czech Republic.,Department of Anesthesiology, Perioperative and Intensive Care Medicine, Faculty of Healthcare Studies, J. E. Purkyne University in Usti nad Labem and Krajska zdravotni a.s. (Regional Healthcare JSC), Masaryk Hospital in Usti nad Labem, Usti nad Labem, Czech Republic.,Center of Research and Development, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic.,Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada.,Technical University in Liberec, Liberec, Czech Republic
| |
Collapse
|
19
|
D'Alessandro R, Meldolesi J. News about non-secretory exocytosis: mechanisms, properties, and functions. J Mol Cell Biol 2020; 11:736-746. [PMID: 30605539 PMCID: PMC6821209 DOI: 10.1093/jmcb/mjy084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 11/14/2018] [Accepted: 01/02/2019] [Indexed: 12/22/2022] Open
Abstract
The fusion by exocytosis of many vesicles to the plasma membrane induces the discharge to the extracellular space of their abundant luminal cargoes. Other exocytic vesicles, however, do not contain cargoes, and thus, their fusion is not followed by secretion. Therefore, two distinct processes of exocytosis exist, one secretory and the other non-secretory. The present review deals with the knowledge of non-secretory exocytosis developed during recent years. Among such developments are the dual generation of the exocytic vesicles, initially released either from the trans-Golgi network or by endocytosis; their traffic with activation of receptors, channels, pumps, and transporters; the identification of their tethering and soluble N-ethylmaleimide-sensitive factor attachment protein receptor complexes that govern membrane fusions; the growth of axons and the membrane repair. Examples of potential relevance of these processes for pathology and medicine are also reported. The developments presented here offer interesting chances for future progress in the field.
Collapse
Affiliation(s)
| | - Jacopo Meldolesi
- Scientific Institute San Raffaele and Vita Salute San Raffaele University, Via Olgettina 58, Milan, Italy
| |
Collapse
|
20
|
Wall SM, Verlander JW, Romero CA. The Renal Physiology of Pendrin-Positive Intercalated Cells. Physiol Rev 2020; 100:1119-1147. [PMID: 32347156 PMCID: PMC7474261 DOI: 10.1152/physrev.00011.2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Intercalated cells (ICs) are found in the connecting tubule and the collecting duct. Of the three IC subtypes identified, type B intercalated cells are one of the best characterized and known to mediate Cl- absorption and HCO3- secretion, largely through the anion exchanger pendrin. This exchanger is thought to act in tandem with the Na+-dependent Cl-/HCO3- exchanger, NDCBE, to mediate net NaCl absorption. Pendrin is stimulated by angiotensin II and aldosterone administration via the angiotensin type 1a and the mineralocorticoid receptors, respectively. It is also stimulated in models of metabolic alkalosis, such as with NaHCO3 administration. In some rodent models, pendrin-mediated HCO3- secretion modulates acid-base balance. However, of probably more physiological or clinical significance is the role of these pendrin-positive ICs in blood pressure regulation, which occurs, at least in part, through pendrin-mediated renal Cl- absorption, as well as their effect on the epithelial Na+ channel, ENaC. Aldosterone stimulates ENaC directly through principal cell mineralocorticoid hormone receptor (ligand) binding and also indirectly through its effect on pendrin expression and function. In so doing, pendrin contributes to the aldosterone pressor response. Pendrin may also modulate blood pressure in part through its action in the adrenal medulla, where it modulates the release of catecholamines, or through an indirect effect on vascular contractile force. In addition to its role in Na+ and Cl- balance, pendrin affects the balance of other ions, such as K+ and I-. This review describes how aldosterone and angiotensin II-induced signaling regulate pendrin and the contribution of pendrin-positive ICs in the kidney to distal nephron function and blood pressure.
Collapse
Affiliation(s)
- Susan M Wall
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Jill W Verlander
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Cesar A Romero
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
21
|
Touré A. Importance of SLC26 Transmembrane Anion Exchangers in Sperm Post-testicular Maturation and Fertilization Potential. Front Cell Dev Biol 2019; 7:230. [PMID: 31681763 PMCID: PMC6813192 DOI: 10.3389/fcell.2019.00230] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/26/2019] [Indexed: 12/17/2022] Open
Abstract
In mammals, sperm cells produced within the testis are structurally differentiated but remain immotile and are unable to fertilize the oocyte unless they undergo a series of maturation events during their transit in the male and female genital tracts. This post-testicular functional maturation is known to rely on the micro-environment of both male and female genital tracts, and is tightly controlled by the pH of their luminal milieus. In particular, within the epididymis, the establishment of a low bicarbonate (HCO3–) concentration contributes to luminal acidification, which is necessary for sperm maturation and subsequent storage in a quiescent state. Following ejaculation, sperm is exposed to the basic pH of the female genital tract and bicarbonate (HCO3–), calcium (Ca2+), and chloride (Cl–) influxes induce biochemical and electrophysiological changes to the sperm cells (cytoplasmic alkalinization, increased cAMP concentration, and protein phosphorylation cascades), which are indispensable for the acquisition of fertilization potential, a process called capacitation. Solute carrier 26 (SLC26) members are conserved membranous proteins that mediate the transport of various anions across the plasma membrane of epithelial cells and constitute important regulators of pH and HCO3– concentration. Most SLC26 members were shown to physically interact and cooperate with the cystic fibrosis transmembrane conductance regulator channel (CFTR) in various epithelia, mainly by stimulating its Cl– channel activity. Among SLC26 members, the function of SLC26A3, A6, and A8 were particularly investigated in the male genital tract and the sperm cells. In this review, we will focus on SLC26s contributions to ionic- and pH-dependent processes during sperm post-testicular maturation. We will specify the current knowledge regarding their functions, based on data from the literature generated by means of in vitro and in vivo studies in knock-out mouse models together with genetic studies of infertile patients. We will also discuss the limits of those studies, the current research gaps and identify some key points for potential developments in this field.
Collapse
Affiliation(s)
- Aminata Touré
- INSERM U1016, Centre National de la Recherche Scientifique, UMR 8104, Institut Cochin, Université de Paris, Paris, France
| |
Collapse
|
22
|
Abstract
The epithelium of the kidney collecting duct (CD) is composed mainly of two different types of cells with distinct and complementary functions. CD principal cells traditionally have been considered to have a major role in Na+ and water regulation, while intercalated cells (ICs) were thought to largely modulate acid-base homeostasis. In recent years, our understanding of IC function has improved significantly owing to new research findings. Thus, we now have a new model for CD transport that integrates mechanisms of salt and water reabsorption, K+ homeostasis, and acid-base status between principal cells and ICs. There are three main types of ICs (type A, type B, and non-A, non-B), which first appear in the late distal convoluted tubule or in the connecting segment in a species-dependent manner. ICs can be detected in CD from cortex to the initial part of the inner medulla, although some transport proteins that are key components of ICs also are present in medullary CD, cells considered inner medullary. Of the three types of ICs, each has a distinct morphology and expresses different complements of membrane transport proteins that translate into very different functions in homeostasis and contributions to CD luminal pro-urine composition. This review includes recent discoveries in IC intracellular and paracrine signaling that contributes to acid-base regulation as well as Na+, Cl-, K+, and Ca2+ homeostasis. Thus, these new findings highlight the potential role of ICs as targets for potential hypertension treatments.
Collapse
Affiliation(s)
- Renee Rao
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Núria M Pastor-Soler
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA.
| |
Collapse
|
23
|
Seidler U, Nikolovska K. Slc26 Family of Anion Transporters in the Gastrointestinal Tract: Expression, Function, Regulation, and Role in Disease. Compr Physiol 2019; 9:839-872. [DOI: 10.1002/cphy.c180027] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Bissler JJ, Zadjali F, Bridges D, Astrinidis A, Barone S, Yao Y, Redd JR, Siroky BJ, Wang Y, Finley JT, Rusiniak ME, Baumann H, Zahedi K, Gross KW, Soleimani M. Tuberous sclerosis complex exhibits a new renal cystogenic mechanism. Physiol Rep 2019; 7:e13983. [PMID: 30675765 PMCID: PMC6344348 DOI: 10.14814/phy2.13983] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a tumor predisposition syndrome with significant renal cystic and solid tumor disease. While the most common renal tumor in TSC, the angiomyolipoma, exhibits a loss of heterozygosity associated with disease, we have discovered that the renal cystic epithelium is composed of type A intercalated cells that have an intact Tsc gene that have been induced to exhibit Tsc-mutant disease phenotype. This mechanism appears to be different than that for ADPKD. The murine models described here closely resemble the human disease and both appear to be mTORC1 inhibitor responsive. The induction signaling driving cystogenesis may be mediated by extracellular vesicle trafficking.
Collapse
Affiliation(s)
- John J. Bissler
- Department of PediatricsUniversity of Tennessee Health Science Center and Le Bonheur Children's HospitalMemphisTennessee
- St. Jude Children's Research HospitalMemphisTennessee
| | - Fahad Zadjali
- Department of Clinical BiochemistryCollege of Medicine & Health SciencesSultan Qaboos UniversityMuscatOman
| | - Dave Bridges
- Department of Nutritional SciencesUniversity of Michigan School of Public HealthAnn ArborMichigan
| | - Aristotelis Astrinidis
- Department of PediatricsUniversity of Tennessee Health Science Center and Le Bonheur Children's HospitalMemphisTennessee
| | - Sharon Barone
- Departments of MedicineUniversity of Cincinnati College of MedicineCincinnatiOhio
- Center on Genetics of TransportUniversity of Cincinnati College of MedicineCincinnatiOhio
- Research ServicesVeterans Affairs Medical CenterCincinnatiOhio
| | - Ying Yao
- Department of PediatricsUniversity of Tennessee Health Science Center and Le Bonheur Children's HospitalMemphisTennessee
| | - JeAnna R. Redd
- Department of Nutritional SciencesUniversity of Michigan School of Public HealthAnn ArborMichigan
| | - Brian J. Siroky
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhio
| | - Yanqing Wang
- Department of Molecular and Cellular BiologyRoswell Park Cancer InstituteBuffaloNew York
| | - Joel T. Finley
- Department of PediatricsUniversity of Tennessee Health Science Center and Le Bonheur Children's HospitalMemphisTennessee
| | - Michael E. Rusiniak
- Department of Molecular and Cellular BiologyRoswell Park Cancer InstituteBuffaloNew York
| | - Heinz Baumann
- Department of Molecular and Cellular BiologyRoswell Park Cancer InstituteBuffaloNew York
| | - Kamyar Zahedi
- Departments of MedicineUniversity of Cincinnati College of MedicineCincinnatiOhio
- Center on Genetics of TransportUniversity of Cincinnati College of MedicineCincinnatiOhio
- Research ServicesVeterans Affairs Medical CenterCincinnatiOhio
| | - Kenneth W. Gross
- Department of Molecular and Cellular BiologyRoswell Park Cancer InstituteBuffaloNew York
| | - Manoocher Soleimani
- Departments of MedicineUniversity of Cincinnati College of MedicineCincinnatiOhio
- Center on Genetics of TransportUniversity of Cincinnati College of MedicineCincinnatiOhio
- Research ServicesVeterans Affairs Medical CenterCincinnatiOhio
| |
Collapse
|
25
|
Barone S, Xu J, Zahedi K, Brooks M, Soleimani M. Probenecid Pre-treatment Downregulates the Kidney Cl -/HCO 3- Exchanger (Pendrin) and Potentiates Hydrochlorothiazide-Induced Diuresis. Front Physiol 2018; 9:849. [PMID: 30050451 PMCID: PMC6050369 DOI: 10.3389/fphys.2018.00849] [Citation(s) in RCA: 7] [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: 03/16/2018] [Accepted: 06/14/2018] [Indexed: 01/14/2023] Open
Abstract
Background: Probenecid is a uricosuric agent that in addition to exerting a positive ionotropic effect in the heart, blocks the ATP transporter Pannexin 1 and inhibits the Cl-/HCO3- exchanger, pendrin. In the kidney, pendrin blunts the loss of salt wasting secondary to the inhibition of the thiazide-sensitive Na+-Cl- co-transporter (NCC/SLC12A3). Hypothesis: Pre-treatment with probenecid down-regulates pendrin; therefore, leaving NCC as the main salt absorbing transporter in the distal nephron, and hence enhances the hydrochlorothiazide (HCTZ)-induced diuresis. Methods: Daily balance studies, blood and urine chemical analysis, immunofluorescence, as well as western and northern blot analyses were utilized to examine the effects of probenecid alone (at 250 mg/kg/day) or in combination with HCTZ (at 40 mg/kg/day) on kidney function and on salt and water transporters in the collecting duct. Results: Male Sprague Dawley rats were subjected to three different protocols: (1) HCTZ for 4 days, (2) probenecid for 10 days, and (3) primed with probenecid for 6 days followed by probenecid and HCTZ for 4 additional days. Treatment protocol 1 (HCTZ for 4 days) only mildly increased the urine volume (U Vol) from a baseline of 9.8-13.4 ml/day. In response to treatment protocol 2 (probenecid for 10 days), U Vol increased to 15.9 ml/24 h. Treatment protocol 3 (probenecid for 6 days followed by probenecid and HCTZ for 4 additional days) increased the U Vol to 42.9 ml/day on day 4 of co-treatment with HCTZ and probenecid (compared to probenecid p = 0.003, n = 5 or HCTZ alone p = 0.001, n = 5). Probenecid treatment at 250 mg/kg/day downregulated the expression of pendrin and led to a decrease in AQP2 expression. Enhanced diuresis by probenecid plus HCTZ was not associated with volume depletion. Conclusion: Probenecid pre-treatment downregulates pendrin and robustly enhances diuresis by HCTZ-mediated NCC inhibition in kidney.
Collapse
Affiliation(s)
- Sharon Barone
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Research Services, VA Medical Center, Cincinnati, OH, United States
| | - Jie Xu
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Center on Genetics of Transport and Epithelial Biology, University of Cincinnati, Cincinnati, OH, United States
| | - Kamyar Zahedi
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Research Services, VA Medical Center, Cincinnati, OH, United States
| | - Marybeth Brooks
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Manoocher Soleimani
- Department of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Research Services, VA Medical Center, Cincinnati, OH, United States
- Center on Genetics of Transport and Epithelial Biology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
26
|
Kurtz I. Renal Tubular Acidosis: H +/Base and Ammonia Transport Abnormalities and Clinical Syndromes. Adv Chronic Kidney Dis 2018; 25:334-350. [PMID: 30139460 PMCID: PMC6128697 DOI: 10.1053/j.ackd.2018.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Renal tubular acidosis (RTA) represents a group of diseases characterized by (1) a normal anion gap metabolic acidosis; (2) abnormalities in renal HCO3- absorption or new renal HCO3- generation; (3) changes in renal NH4+, Ca2+, K+, and H2O homeostasis; and (4) extrarenal manifestations that provide etiologic diagnostic clues. The focus of this review is to give a general overview of the pathogenesis of the various clinical syndromes causing RTA with a particular emphasis on type I (hypokalemic distal RTA) and type II (proximal) RTA while reviewing their pathogenesis from a physiological "bottom-up" approach. In addition, the factors involved in the generation of metabolic acidosis in both type I and II RTA are reviewed highlighting the importance of altered renal ammonia production/partitioning and new HCO3- generation. Our understanding of the underlying tubular transport and extrarenal abnormalities has significantly improved since the first recognition of RTA as a clinical entity because of significant advances in clinical acid-base chemistry, whole tubule and single-cell H+/base transport, and the molecular characterization of the various transporters and channels that are functionally affected in patients with RTA. Despite these advances, additional studies are needed to address the underlying mechanisms involved in hypokalemia, altered ammonia production/partitioning, hypercalciuria, nephrocalcinosis, cystic abnormalities, and CKD progression in these patients.
Collapse
Affiliation(s)
- Ira Kurtz
- Division of Nephrology, David Geffen School of Medicine, and Brain Research Institute, UCLA, Los Angeles, CA.
| |
Collapse
|
27
|
Nanami M, Pham TD, Kim YH, Yang B, Sutliff RL, Staub O, Klein JD, Lopez-Cayuqueo KI, Chambrey R, Park AY, Wang X, Pech V, Verlander JW, Wall SM. The Role of Intercalated Cell Nedd4-2 in BP Regulation, Ion Transport, and Transporter Expression. J Am Soc Nephrol 2018; 29:1706-1719. [PMID: 29773687 DOI: 10.1681/asn.2017080826] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 03/29/2018] [Indexed: 12/23/2022] Open
Abstract
BackgroundNedd4-2 is an E3 ubiquitin-protein ligase that associates with transport proteins, causing their ubiquitylation, and then internalization and degradation. Previous research has suggested a correlation between Nedd4-2 and BP. In this study, we explored the effect of intercalated cell (IC) Nedd4-2 gene ablation on IC transporter abundance and function and on BP.Methods We generated IC Nedd4-2 knockout mice using Cre-lox technology and produced global pendrin/Nedd4-2 null mice by breeding global Nedd4-2 null (Nedd4-2-/- ) mice with global pendrin null (Slc26a4-/- ) mice. Mice ate a diet with 1%-4% NaCl; BP was measured by tail cuff and radiotelemetry. We measured transepithelial transport of Cl- and total CO2 and transepithelial voltage in cortical collecting ducts perfused in vitro Transporter abundance was detected with immunoblots, immunohistochemistry, and immunogold cytochemistry.Results IC Nedd4-2 gene ablation markedly increased electroneutral Cl-/HCO3- exchange in the cortical collecting duct, although benzamil-, thiazide-, and bafilomycin-sensitive ion flux changed very little. IC Nedd4-2 gene ablation did not increase the abundance of type B IC transporters, such as AE4 (Slc4a9), H+-ATPase, barttin, or the Na+-dependent Cl-/HCO3- exchanger (Slc4a8). However, IC Nedd4-2 gene ablation increased CIC-5 total protein abundance, apical plasma membrane pendrin abundance, and the ratio of pendrin expression on the apical membrane to the cytoplasm. IC Nedd4-2 gene ablation increased BP by approximately 10 mm Hg. Moreover, pendrin gene ablation eliminated the increase in BP observed in global Nedd4-2 knockout mice.Conclusions IC Nedd4-2 regulates Cl-/HCO3- exchange in ICs., Nedd4-2 gene ablation increases BP in part through its action in these cells.
Collapse
Affiliation(s)
| | | | | | - Baoli Yang
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, Iowa
| | | | - Olivier Staub
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland.,National Centre of Competence in Research "Kidney.ch," Zurich, Switzerland
| | | | - Karen I Lopez-Cayuqueo
- Centro de Estudios Cientificos, Valdivia, Chile.,Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Université Paris-Descartes, Paris, France
| | - Regine Chambrey
- Institut National de la Santé et de la Recherche Médicale U1188, Universite de la Reunion, Plateforme Cyclotron Réunion Océan Indien, St. Denis, Ile de la Reunion, France; and
| | | | | | | | - Jill W Verlander
- Renal Division, Department of Medicine, University of Florida at Gainesville, Gainesville, Florida
| | - Susan M Wall
- Renal and .,Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
28
|
Rahmati N, Hoebeek FE, Peter S, De Zeeuw CI. Chloride Homeostasis in Neurons With Special Emphasis on the Olivocerebellar System: Differential Roles for Transporters and Channels. Front Cell Neurosci 2018; 12:101. [PMID: 29765304 PMCID: PMC5938380 DOI: 10.3389/fncel.2018.00101] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
The intraneuronal ionic composition is an important determinant of brain functioning. There is growing evidence that aberrant homeostasis of the intracellular concentration of Cl- ([Cl-]i) evokes, in addition to that of Na+ and Ca2+, robust impairments of neuronal excitability and neurotransmission and thereby neurological conditions. More specifically, understanding the mechanisms underlying regulation of [Cl-]i is crucial for deciphering the variability in GABAergic and glycinergic signaling of neurons, in both health and disease. The homeostatic level of [Cl-]i is determined by various regulatory mechanisms, including those mediated by plasma membrane Cl- channels and transporters. This review focuses on the latest advances in identification, regulation and characterization of Cl- channels and transporters that modulate neuronal excitability and cell volume. By putting special emphasis on neurons of the olivocerebellar system, we establish that Cl- channels and transporters play an indispensable role in determining their [Cl-]i and thereby their function in sensorimotor coordination.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Freek E. Hoebeek
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- NIDOD Institute, Wilhelmina Children's Hospital, University Medical Center Utrecht and Brain Center Rudolf Magnus, Utrecht, Netherlands
| | - Saša Peter
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Dutch Academy for Arts and Sciences, Amsterdam, Netherlands
| |
Collapse
|
29
|
De la Vieja A, Santisteban P. Role of iodide metabolism in physiology and cancer. Endocr Relat Cancer 2018; 25:R225-R245. [PMID: 29437784 DOI: 10.1530/erc-17-0515] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/28/2022]
Abstract
Iodide (I-) metabolism is crucial for the synthesis of thyroid hormones (THs) in the thyroid and the subsequent action of these hormones in the organism. I- is principally transported by the sodium iodide symporter (NIS) and by the anion exchanger PENDRIN, and recent studies have demonstrated the direct participation of new transporters including anoctamin 1 (ANO1), cystic fibrosis transmembrane conductance regulator (CFTR) and sodium multivitamin transporter (SMVT). Several of these transporters have been found expressed in various tissues, implicating them in I- recycling. New research supports the exciting idea that I- participates as a protective antioxidant and can be oxidized to hypoiodite, a potent oxidant involved in the host defense against microorganisms. This was possibly the original role of I- in biological systems, before the appearance of TH in evolution. I- per se participates in its own regulation, and new evidence indicates that it may be antineoplastic, anti-proliferative and cytotoxic in human cancer. Alterations in the expression of I- transporters are associated with tumor development in a cancer-type-dependent manner and, accordingly, NIS, CFTR and ANO1 have been proposed as tumor markers. Radioactive iodide has been the mainstay adjuvant treatment for thyroid cancer for the last seven decades by virtue of its active transport by NIS. The rapid advancement of techniques that detect radioisotopes, in particular I-, has made NIS a preferred target-specific theranostic agent.
Collapse
Affiliation(s)
- Antonio De la Vieja
- Tumor Endocrine Unit, Chronic Disease Program (UFIEC), Instituto de Salud Carlos III, Madrid, Spain
- CiberOnc, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Santisteban
- CiberOnc, Instituto de Salud Carlos III, Madrid, Spain
- Department of Physiopathology of Endocrine a Nervous System, Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| |
Collapse
|
30
|
Tomilin VN, Zaika O, Subramanya AR, Pochynyuk O. Dietary K + and Cl - independently regulate basolateral conductance in principal and intercalated cells of the collecting duct. Pflugers Arch 2018; 470:339-353. [PMID: 29134279 PMCID: PMC9624487 DOI: 10.1007/s00424-017-2084-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/17/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
The renal collecting duct contains two distinct cell types, principal and intercalated cells, expressing potassium Kir4.1/5.1 (KCNJ10/16) and chloride ClC-K2 (ClC-Kb in humans) channels on their basolateral membrane, respectively. Both channels are thought to play important roles in controlling systemic water-electrolyte balance and blood pressure. However, little is known about mechanisms regulating activity of Kir4.1/5.1 and ClC-K2/b. Here, we employed patch clamp analysis at the single channel and whole cell levels in freshly isolated mouse collecting ducts to investigate regulation of Kir4.1/5.1 and ClC-K2/b by dietary K+ and Cl- intake. Treatment of mice with high K+ and high Cl- diet (6% K+, 5% Cl-) for 1 week significantly increased basolateral K+-selective current, single channel Kir4.1/5.1 activity and induced hyperpolarization of basolateral membrane potential in principal cells when compared to values in mice on a regular diet (0.9% K+, 0.5% Cl-). In contrast, basolateral Cl--selective current and single channel ClC-K2/b activity was markedly decreased in intercalated cells under this condition. Substitution of dietary K+ to Na+ in the presence of high Cl- exerted a similar inhibiting action of ClC-K2/b suggesting that the channel is sensitive to variations in dietary Cl- per se. Cl--sensitive with-no-lysine kinase (WNK) cascade has been recently proposed to orchestrate electrolyte transport in the distal tubule during variations of dietary K+. However, co-expression of WNK1 or its major downstream effector Ste20-related proline-alanine-rich kinase (SPAK) had no effect on ClC-Kb over-expressed in Chinese hamster ovary (CHO) cells. Treatment of mice with high K+ diet without concomitant elevations in dietary Cl- (6% K+, 0.5% Cl-) elicited a comparable increase in basolateral K+-selective current, single channel Kir4.1/5.1 activity in principal cells, but had no significant effect on ClC-K2/b activity in intercalated cells. Furthermore, stimulation of aldosterone signaling by Deoxycorticosterone acetate (DOCA) recapitulated the stimulatory actions of high K+ intake on Kir4.1/5.1 channels in principal cells but was ineffective to alter ClC-K2/b activity and basolateral Cl- conductance in intercalated cells. In summary, we report that variations of dietary K+ and Cl- independently regulate basolateral potassium and chloride conductance in principal and intercalated cells. We propose that such discrete mechanism might contribute to fine-tuning of urinary excretion of electrolytes depending on dietary intake.
Collapse
Affiliation(s)
- Viktor N Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX, 77030, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX, 77030, USA
| | - Arohan R Subramanya
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX, 77030, USA.
| |
Collapse
|
31
|
Edwards A, Crambert G. Versatility of NaCl transport mechanisms in the cortical collecting duct. Am J Physiol Renal Physiol 2017; 313:F1254-F1263. [PMID: 28877883 DOI: 10.1152/ajprenal.00369.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/16/2017] [Accepted: 08/31/2017] [Indexed: 12/16/2022] Open
Abstract
The cortical collecting duct (CCD) forms part of the aldosterone-sensitive distal nephron and plays an essential role in maintaining the NaCl balance and acid-base status. The CCD epithelium comprises principal cells as well as different types of intercalated cells. Until recently, transcellular Na+ transport was thought to be restricted to principal cells, whereas (acid-secreting) type A and (bicarbonate-secreting) type B intercalated cells were associated with the regulation of acid-base homeostasis. This review describes how this traditional view has been upended by several discoveries in the past decade. A series of studies has shown that type B intercalated cells can mediate electroneutral NaCl reabsorption by a mechanism involving Na+-dependent and Na+-independent Cl-/[Formula: see text] exchange, and that is energetically driven by basolateral vacuolar H+-ATPase pumps. Other research indicates that type A intercalated cells can mediate NaCl secretion, through a bumetanide-sensitive pathway that is energized by apical H+,K+-ATPase type 2 pumps operating as Na+/K+ exchangers. We also review recent findings on the contribution of the paracellular route to NaCl transport in the CCD. Last, we describe cross-talk processes, by which one CCD cell type impacts Na+/Cl- transport in another cell type. The mechanisms that have been identified to date demonstrate clearly the interdependence of NaCl and acid-base transport systems in the CCD. They also highlight the remarkable versatility of this nephron segment.
Collapse
Affiliation(s)
- Aurélie Edwards
- Sorbonne Universités, UPMC Univ Paris 06, Université Paris Descartes, Sorbonne Paris Cité, INSERM UMRS 1138, CNRS ERL 8228, Centre de Recherche des Cordeliers, Paris, France; and .,Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Gilles Crambert
- Sorbonne Universités, UPMC Univ Paris 06, Université Paris Descartes, Sorbonne Paris Cité, INSERM UMRS 1138, CNRS ERL 8228, Centre de Recherche des Cordeliers, Paris, France; and
| |
Collapse
|
32
|
MIST1 Links Secretion and Stress as both Target and Regulator of the Unfolded Protein Response. Mol Cell Biol 2016; 36:2931-2944. [PMID: 27644325 DOI: 10.1128/mcb.00366-16] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 07/22/2016] [Accepted: 09/10/2016] [Indexed: 12/28/2022] Open
Abstract
Transcriptional networks that govern secretory cell specialization, including instructing cells to develop a unique cytoarchitecture, amass extensive protein synthesis machinery, and be embodied to respond to endoplasmic reticulum (ER) stress, remain largely uncharacterized. In this study, we discovered that the secretory cell transcription factor MIST1 (Bhlha15), previously shown to be essential for cytoskeletal organization and secretory activity, also functions as a potent ER stress-inducible transcriptional regulator. Genome-wide DNA binding studies, coupled with genetic mouse models, revealed MIST1 gene targets that function along the entire breadth of the protein synthesis, processing, transport, and exocytosis networks. Additionally, key MIST1 targets are essential for alleviating ER stress in these highly specialized cells. Indeed, MIST1 functions as a coregulator of the unfolded protein response (UPR) master transcription factor XBP1 for a portion of target genes that contain adjacent MIST1 and XBP1 binding sites. Interestingly, Mist1 gene expression is induced during ER stress by XBP1, but as ER stress subsides, MIST1 serves as a feedback inhibitor, directly binding the Xbp1 promoter and repressing Xbp1 transcript production. Together, our findings provide a new paradigm for XBP1-dependent UPR regulation and position MIST1 as a potential biotherapeutic for numerous human diseases.
Collapse
|
33
|
Morla L, Doucet A, Lamouroux C, Crambert G, Edwards A. The renal cortical collecting duct: a secreting epithelium? J Physiol 2016; 594:5991-6008. [PMID: 27412964 PMCID: PMC5063930 DOI: 10.1113/jp272877] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/07/2016] [Indexed: 01/01/2023] Open
Abstract
KEY POINTS The cortical collecting duct (CCD) plays an essential role in sodium homeostasis by fine-tuning the amount of sodium that is excreted in the urine. Ex vivo, the microperfused CCD reabsorbs sodium in the absence of lumen-to-bath concentration gradients. In the present study, we show that, in the presence of physiological lumen-to-bath concentration gradients, and in the absence of endocrine, paracrine and neural regulation, the mouse CCD secretes sodium, which represents a paradigm shift. This secretion occurs via the paracellular route, as well as a transcellular pathway that is energized by apical H+ /K+ -ATPase type 2 pumps operating as Na+ /K+ exchangers. The newly identified transcellular secretory pathway represents a physiological target for the regulation of sodium handling and for anti-hypertensive therapeutic agents. ABSTRACT In vitro microperfusion experiments have demonstrated that cortical collecting ducts (CCDs) reabsorb sodium via principal and type B intercalated cells under sodium-depleted conditions and thereby contribute to sodium and blood pressure homeostasis. However, these experiments were performed in the absence of the transepithelial ion concentration gradients that prevail in vivo and determine paracellular transport. The present study aimed to characterize Na+ , K+ and Cl- fluxes in the mouse CCD in the presence of physiological transepithelial concentration gradients. For this purpose, we combined in vitro measurements of ion fluxes across microperfused CCDs of sodium-depleted mice with the predictions of a mathematical model. When NaCl transport was inhibited in all cells, CCDs secreted Na+ and reabsorbed K+ ; Cl- transport was negligible. Removing inhibitors of type A and B intercalated cells increased Na+ secretion in wild-type (WT) mice but not in H+ /K+ -ATPase type 2 (HKA2) knockout mice. Further inhibition of basolateral NaCl entry via the Na+ -K+ -2Cl- cotransporter in type A intercalated cells reduced Na+ secretion in WT mice to the levels observed in HKA2-/- mice. With no inhibitors, WT mouse CCDs still secreted Na+ and reabsorbed K+ . In vivo, HKA2-/- mice excreted less Na+ than WT mice after switching to a high-salt diet. Taken together, our results indicate that type A intercalated cells secrete Na+ via basolateral Na+ -K+ -2Cl- cotransporters in tandem with apical HKA2 pumps. They also suggest that the CCD can mediate overall Na+ secretion, and that its ability to reabsorb NaCl in vivo depends on the presence of acute regulatory factors.
Collapse
Affiliation(s)
- Luciana Morla
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, CNRS, ERL 8228, Centre de Recherche des Cordeliers, Paris, France
| | - Alain Doucet
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, CNRS, ERL 8228, Centre de Recherche des Cordeliers, Paris, France
| | - Christine Lamouroux
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, CNRS, ERL 8228, Centre de Recherche des Cordeliers, Paris, France
| | | | | |
Collapse
|
34
|
SLC26A11 (KBAT) in Purkinje Cells Is Critical for Inhibitory Transmission and Contributes to Locomotor Coordination. eNeuro 2016; 3:eN-NWR-0028-16. [PMID: 27390771 PMCID: PMC4908300 DOI: 10.1523/eneuro.0028-16.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/21/2016] [Accepted: 05/24/2016] [Indexed: 11/24/2022] Open
Abstract
Chloride homeostasis determines the impact of inhibitory synaptic transmission and thereby mediates the excitability of neurons. Even though cerebellar Purkinje cells (PCs) receive a pronounced inhibitory GABAergic input from stellate and basket cells, the role of chloride homeostasis in these neurons is largely unknown. Here we studied at both the cellular and systems physiological level the function of a recently discovered chloride channel, SLC26A11 or kidney brain anion transporter (KBAT), which is prominently expressed in PCs. Using perforated patch clamp recordings of PCs, we found that a lack of KBAT channel in PC-specific KBAT KO mice (L7-KBAT KOs) induces a negative shift in the reversal potential of chloride as reflected in the GABAA-receptor-evoked currents, indicating a decrease in intracellular chloride concentration. Surprisingly, both in vitro and in vivo PCs in L7-KBAT KOs showed a significantly increased action potential firing frequency of simple spikes, which correlated with impaired motor performance on the Erasmus Ladder. Our findings support an important role for SLC26A11 in moderating chloride homeostasis and neuronal activity in the cerebellum.
Collapse
|
35
|
Zaika O, Tomilin V, Mamenko M, Bhalla V, Pochynyuk O. New perspective of ClC-Kb/2 Cl- channel physiology in the distal renal tubule. Am J Physiol Renal Physiol 2016; 310:F923-30. [PMID: 26792067 PMCID: PMC5002062 DOI: 10.1152/ajprenal.00577.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/14/2016] [Indexed: 12/17/2022] Open
Abstract
Since its identification as the underlying molecular cause of Bartter's syndrome type 3, ClC-Kb (ClC-K2 in rodents, henceforth it will be referred as ClC-Kb/2) is proposed to play an important role in systemic electrolyte balance and blood pressure regulation by controlling basolateral Cl(-) exit in the distal renal tubular segments from the cortical thick ascending limb to the outer medullary collecting duct. Considerable experimental and clinical effort has been devoted to the identification and characterization of disease-causing mutations as well as control of the channel by its cofactor, barttin. However, we have only begun to unravel the role of ClC-Kb/2 in different tubular segments and to reveal the regulators of its expression and function, e.g., insulin and IGF-1. In this review we discuss recent experimental evidence in this regard and highlight unexplored questions critical to understanding ClC-Kb/2 physiology in the kidney.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Viktor Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University, Stanford, California
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas; and
| |
Collapse
|
36
|
Hosoya M, Fujioka M, Kobayashi R, Okano H, Ogawa K. Overlapping expression of anion exchangers in the cochlea of a non-human primate suggests functional compensation. Neurosci Res 2016; 110:1-10. [PMID: 27091614 DOI: 10.1016/j.neures.2016.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/27/2016] [Accepted: 04/05/2016] [Indexed: 11/19/2022]
Abstract
Ion homeostasis in the inner ear is essential for proper hearing. Anion exchangers are one of the transporters responsible for the maintenance of homeostasis, but their expression profile in the primate cochlea has not been fully characterized. However, species-specific overlapping expression patterns and functional compensation in other organs, such as the kidney, pancreas and small intestine, have been reported. Here, we determined the expression patterns of the anion exchangers SLC26A4, SLC26A5, SLC26A6, SLC26A7, SLC26A11, SLC4A2 and SLC4A3 in the cochlea of a non-human primate, the common marmoset (Callithrix jacchus). Although the pattern of expression of SLC26A4 and SLC26A5 was similar to that in rodents, SLC26A7, SLC4A2, SLC4A3 exhibited different distributions. Notably, five transporters, SLC26A4, SLC26A6, SLC26A11 SLC4A2 and SLC4A3, were expressed in the cells of the outer sulcus. Our results reveal a species-specific distribution pattern of anion exchangers in the cochlea, particularly in the outer sulcus cells, suggesting functional compensation among these exchangers. This "primate-specific" pattern may be related to the human-specific hearing loss phenotypes of channelopathy disorders, including the SLC26A4-related diseases Pendred syndrome/DFNB4.
Collapse
Affiliation(s)
- Makoto Hosoya
- Keio University School of Medicine, Department of Otorhinolaryngology, Head and Neck Surgery, 35 Shinanomachi, Shinjuku-ku 160-8582, Japan
| | - Masato Fujioka
- Keio University School of Medicine, Department of Otorhinolaryngology, Head and Neck Surgery, 35 Shinanomachi, Shinjuku-ku 160-8582, Japan.
| | - Reona Kobayashi
- Keio University School of Medicine, Department of Physiology, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Keio University School of Medicine, Department of Physiology, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kaoru Ogawa
- Keio University School of Medicine, Department of Otorhinolaryngology, Head and Neck Surgery, 35 Shinanomachi, Shinjuku-ku 160-8582, Japan
| |
Collapse
|
37
|
The Role of Epithelial Sodium Channel ENaC and the Apical Cl-/HCO3- Exchanger Pendrin in Compensatory Salt Reabsorption in the Setting of Na-Cl Cotransporter (NCC) Inactivation. PLoS One 2016; 11:e0150918. [PMID: 26963391 PMCID: PMC4786216 DOI: 10.1371/journal.pone.0150918] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/18/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The absence of NCC does not cause significant salt wasting in NCC deficient mice under basal conditions. We hypothesized that ENaC and pendrin play important roles in compensatory salt absorption in the setting of NCC inactivation, and their inhibition and/or downregulation can cause significant salt wasting in NCC KO mice. METHODS WT and NCC KO mice were treated with a daily injection of either amiloride, an inhibitor of ENaC, or acetazolamide (ACTZ), a blocker of salt and bicarbonate reabsorption in the proximal tubule and an inhibitor of carbonic anhydrases in proximal tubule and intercalated cells, or a combination of acetazolamide plus amiloride for defined durations. Animals were subjected to daily balance studies. At the end of treatment, kidneys were harvested and examined. Blood samples were collected for electrolytes and acid base analysis. RESULTS Amiloride injection significantly increased the urine output (UO) in NCC KO mice (from 1.3 ml/day before to 2.5 ml/day after amiloride, p<0.03, n = 4) but caused only a slight change in UO in WT mice (p>0.05). The increase in UO in NCC KO mice was associated with a significant increase in sodium excretion (from 0.25 mmol/24 hrs at baseline to 0.35 mmol/24 hrs after amiloride injection, p<0.05, n = 4). Daily treatment with ACTZ for 6 days resulted in >80% reduction of kidney pendrin expression in both WT and NCC KO mice. However, ACTZ treatment noticeably increased urine output and salt excretion only in NCC KO mice (with urine output increasing from a baseline of 1.1 ml/day to 2.3 ml/day and sodium excretion increasing from 0.22 mmole/day before to 0.31 mmole/day after ACTZ) in NCC KO mice; both parameters were significantly higher than in WT mice. Western blot analysis demonstrated significant enhancement in ENaC expression in medulla and cortex of NCC KO and WT mice in response to ACTZ injection for 6 days, and treatment with amiloride in ACTZ-pretreated mice caused a robust increase in salt excretion in both NCC KO and WT mice. Pendrin KO mice did not display a significant increase in urine output or salt excretion after treatment with amiloride or ACTZ. CONCLUSION 1. ENaC plays an important role in salt reabsorption in NCC KO mice. 2. NCC contributes to compensatory salt reabsorption in the setting of carbonic anhydrase inhibition, which is associated with increased delivery of salt from the proximal tubule and the down regulation of pendrin. 3. ENaC is upregulated by ACTZ treatment and its inhibition by amiloride causes significant diuresis in NCC KO and WT mice. Despite being considered mild agents individually, we propose that the combination of acetazolamide and amiloride in the setting of NCC inhibition (i.e., hydrochlorothiazide) will be a powerful diuretic regimen.
Collapse
|
38
|
Nanami M, Lazo-Fernandez Y, Pech V, Verlander JW, Agazatian D, Weinstein AM, Bao HF, Eaton DC, Wall SM. ENaC inhibition stimulates HCl secretion in the mouse cortical collecting duct. I. Stilbene-sensitive Cl- secretion. Am J Physiol Renal Physiol 2015; 309:F251-8. [PMID: 25925258 DOI: 10.1152/ajprenal.00471.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 04/27/2015] [Indexed: 11/22/2022] Open
Abstract
Inhibition of the epithelial Na(+) channel (ENaC) reduces Cl(-) absorption in cortical collecting ducts (CCDs) from aldosterone-treated rats and mice. Since ENaC does not transport Cl(-), the purpose of the present study was to explore how ENaC modulates Cl(-) absorption in mouse CCDs perfused in vitro. Therefore, we measured transepithelial Cl(-) flux and transepithelial voltage in CCDs perfused in vitro taken from mice that consumed a NaCl-replete diet alone or the diet with aldosterone administered by minipump. We observed that application of an ENaC inhibitor [benzamil (3 μM)] to the luminal fluid unmasks conductive Cl(-) secretion. During ENaC blockade, this Cl(-) secretion fell with the application of a nonselective Cl(-) channel blocker [DIDS (100 μM)] to the perfusate. While single channel recordings of intercalated cell apical membranes in split-open CCDs demonstrated a Cl(-) channel with properties that resemble the ClC family of Cl(-) channels, ClC-5 is not the primary pathway for benzamil-sensitive Cl(-) flux. In conclusion, first, in CCDs from aldosterone-treated mice, most Cl(-) absorption is benzamil sensitive, and, second, benzamil application stimulates stilbene-sensitive conductive Cl(-) secretion, which occurs through a ClC-5-independent pathway.
Collapse
Affiliation(s)
- Masayoshi Nanami
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | | | - Vladimir Pech
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jill W Verlander
- Department of Medicine, The University of Florida, Gainesville, Florida
| | - Diana Agazatian
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, Ithaca, New York; Department of Medicine, Weill Medical College of Cornell University, Ithaca, New York; and
| | - Hui-Fang Bao
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Douglas C Eaton
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Susan M Wall
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Physiology, Emory University School of Medicine, Atlanta, Georgia;
| |
Collapse
|
39
|
Rungta R, Choi H, Tyson J, Malik A, Dissing-Olesen L, Lin P, Cain S, Cullis P, Snutch T, MacVicar B. The Cellular Mechanisms of Neuronal Swelling Underlying Cytotoxic Edema. Cell 2015; 161:610-621. [DOI: 10.1016/j.cell.2015.03.029] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/03/2015] [Accepted: 03/12/2015] [Indexed: 01/08/2023]
|
40
|
Roy A, Al-bataineh MM, Pastor-Soler NM. Collecting duct intercalated cell function and regulation. Clin J Am Soc Nephrol 2015; 10:305-24. [PMID: 25632105 DOI: 10.2215/cjn.08880914] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Intercalated cells are kidney tubule epithelial cells with important roles in the regulation of acid-base homeostasis. However, in recent years the understanding of the function of the intercalated cell has become greatly enhanced and has shaped a new model for how the distal segments of the kidney tubule integrate salt and water reabsorption, potassium homeostasis, and acid-base status. These cells appear in the late distal convoluted tubule or in the connecting segment, depending on the species. They are most abundant in the collecting duct, where they can be detected all the way from the cortex to the initial part of the inner medulla. Intercalated cells are interspersed among the more numerous segment-specific principal cells. There are three types of intercalated cells, each having distinct structures and expressing different ensembles of transport proteins that translate into very different functions in the processing of the urine. This review includes recent findings on how intercalated cells regulate their intracellular milieu and contribute to acid-base regulation and sodium, chloride, and potassium homeostasis, thus highlighting their potential role as targets for the treatment of hypertension. Their novel regulation by paracrine signals in the collecting duct is also discussed. Finally, this article addresses their role as part of the innate immune system of the kidney tubule.
Collapse
Affiliation(s)
- Ankita Roy
- Renal-Electrolyte Division, Department of Medicine; and
| | | | - Núria M Pastor-Soler
- Renal-Electrolyte Division, Department of Medicine; and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania A.R. and M.M.A. contributed equally to this work.
| |
Collapse
|
41
|
Zaika O, Mamenko M, Boukelmoune N, Pochynyuk O. IGF-1 and insulin exert opposite actions on ClC-K2 activity in the cortical collecting ducts. Am J Physiol Renal Physiol 2015; 308:F39-F48. [PMID: 25339702 PMCID: PMC4281695 DOI: 10.1152/ajprenal.00545.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/16/2014] [Indexed: 01/07/2023] Open
Abstract
Despite similar stimulatory actions on the epithelial sodium channel (ENaC)-mediated sodium reabsorption in the distal tubule, insulin promotes kaliuresis, whereas insulin-like growth factor-1 (IGF-1) causes a reduction in urinary potassium levels. The factors contributing to this phenomenon remain elusive. Electrogenic distal nephron ENaC-mediated Na(+) transport establishes driving force for Cl(-) reabsorption and K(+) secretion. Using patch-clamp electrophysiology, we document that a Cl(-) channel is highly abundant on the basolateral plasma membrane of intercalated cells in freshly isolated mouse cortical collecting duct (CCD) cells. The channel has characteristics attributable to the ClC-K2: slow gating kinetics, conductance ∼10 pS, voltage independence, Cl(-)>NO3 (-) anion selectivity, and inhibition/activation by low/high pH, respectively. IGF-1 (100 and 500 nM) acutely stimulates ClC-K2 activity in a reversible manner. Inhibition of PI3-kinase (PI3-K) with LY294002 (20 μM) abrogates activation of ClC-K2 by IGF-1. Interestingly, insulin (100 nM) reversibly decreases ClC-K2 activity in CCD cells. This inhibitory action is independent of PI3-K and is mediated by stimulation of a mitogen-activated protein kinase-dependent cascade. We propose that IGF-1, by stimulating ClC-K2 channels, promotes net Na(+) and Cl(-) reabsorption, thus reducing driving force for potassium secretion by the CCD. In contrast, inhibition of ClC-K2 by insulin favors coupling of Na(+) reabsorption with K(+) secretion at the apical membrane contributing to kaliuresis.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Nabila Boukelmoune
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
42
|
Soleimani M. The multiple roles of pendrin in the kidney. Nephrol Dial Transplant 2014; 30:1257-66. [PMID: 25281699 DOI: 10.1093/ndt/gfu307] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/25/2014] [Indexed: 12/30/2022] Open
Abstract
The [Formula: see text] exchanger pendrin (SLC26A4, PDS) is located on the apical membrane of B-intercalated cells in the kidney cortical collecting duct and the connecting tubules and mediates the secretion of bicarbonate and the reabsorption of chloride. Given its dual function of bicarbonate secretion and chloride reabsorption in the distal tubules, it was thought that pendrin plays important roles in systemic acid-base balance and electrolyte and vascular volume homeostasis under basal conditions. Mice with the genetic deletion of pendrin or humans with inactivating mutations in PDS gene, however, do not display excessive salt and fluid wasting or altered blood pressure under baseline conditions. Very recent reports have unmasked the basis of incongruity between the mild phenotype in mutant mice and the role of pendrin as an important player in salt reabsorption in the distal tubule. These studies demonstrate that pendrin and the Na-Cl cotransporter (NCC; SLC12A3) cross compensate for the loss of each other, therefore masking the role that each transporter plays in salt reabsorption under baseline conditions. In addition, pendrin regulates calcium reabsorption in the distal tubules. Furthermore, combined deletion of pendrin and NCC not only causes severe volume depletion but also results in profound calcium wasting and luminal calcification in medullary collecting ducts. Based on studies in pathophysiological states and the examination of genetically engineered mouse models, the evolving picture points to important roles for pendrin (SLC26A4) in kidney physiology and in disease states. This review summarizes recent advances in the characterization of pendrin and the multiple roles it plays in the kidney, with emphasis on its essential roles in several diverse physiological processes, including chloride homeostasis, vascular volume and blood pressure regulation, calcium excretion and kidney stone formation.
Collapse
Affiliation(s)
- Manoocher Soleimani
- Center on Genetics of Transport and Epithelial Biology, University of Cincinnati, Cincinnati, OH, USA Research Services, Veterans Affairs Medical Center, Cincinnati, OH, USA Department of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
43
|
Alka K, Casey JR. Bicarbonate transport in health and disease. IUBMB Life 2014; 66:596-615. [PMID: 25270914 DOI: 10.1002/iub.1315] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/10/2014] [Indexed: 12/28/2022]
Abstract
Bicarbonate (HCO3(-)) has a central place in human physiology as the waste product of mitochondrial energy production and for its role in pH buffering throughout the body. Because bicarbonate is impermeable to membranes, bicarbonate transport proteins are necessary to enable control of bicarbonate levels across membranes. In humans, 14 bicarbonate transport proteins, members of the SLC4 and SLC26 families, function by differing transport mechanisms. In addition, some anion channels and ZIP metal transporters contribute to bicarbonate movement across membranes. Defective bicarbonate transport leads to diseases, including systemic acidosis, brain dysfunction, kidney stones, and hypertension. Altered expression levels of bicarbonate transporters in patients with breast, colon, and lung cancer suggest an important role of these transporters in cancer.
Collapse
Affiliation(s)
- Kumari Alka
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
44
|
Rahmati N, Kunzelmann K, Xu J, Barone S, Sirianant L, De Zeeuw CI, Soleimani M. Slc26a11 is prominently expressed in the brain and functions as a chloride channel: expression in Purkinje cells and stimulation of V H⁺-ATPase. Pflugers Arch 2013; 465:1583-97. [PMID: 23733100 PMCID: PMC11708839 DOI: 10.1007/s00424-013-1300-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/13/2013] [Accepted: 05/14/2013] [Indexed: 02/07/2023]
Abstract
SLC26A11 (human)/Slc26a11 (mouse), also known as kidney brain anion transporter (KBAT), is a member of the SLC26 anion transporter family and shows abundant mRNA expression in the brain. However, its exact cellular distribution and subcellular localization in the brain and its functional identity and possible physiological roles remain unknown. Expression and immunostaining studies demonstrated that Slc26a11 is abundantly expressed in the cerebellum, with a predominant expression in Purkinje cells. Lower expression levels were detected in hippocampus, olfactory bulb, cerebral cortex, and subcortical structures. Patch clamp studies in HEK293 cells transfected with mouse cDNA demonstrated that Slc26a11 can function as a chloride channel that is active under basal conditions and is not regulated by calcium, forskolin, or co-expression with cystic fibrosis transmembrane regulator. Single and double immunofluorescent labeling studies demonstrated the localization of vacuolar (V) H⁺-ATPase and Slc26a11 (KBAT) in the plasma membrane in Purkinje cells. Functional studies in HEK293 cells indicated that transfection with Slc26a11 stimulated acid transport via endogenous V H⁺-ATPase. We conclude that Slc26a11 (KBAT) is prominently distributed in output neurons of various subcortical and cortical structures in the central nervous system, with specific expression in Purkinje cells and that it may operate as a chloride channel regulating acid translocation by H⁺-ATPase across the plasma membrane and in intracellular compartments.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neuroscience, Erasmus University, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
45
|
Soleimani M. SLC26 Cl-/HCO3- exchangers in the kidney: roles in health and disease. Kidney Int 2013; 84:657-66. [PMID: 23636174 PMCID: PMC10947778 DOI: 10.1038/ki.2013.138] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/25/2013] [Accepted: 02/14/2013] [Indexed: 12/30/2022]
Abstract
Solute-linked carrier 26 (SLC26) isoforms constitute a conserved family of anion transporters with 10 distinct members. Except for SLC26A5 (prestin), all can operate as multifunctional anion exchangers, with three members (SLC26A7, SLC26A9, and SLC26A11) also capable of functioning as chloride channels. Several SLC26 isoforms can specifically mediate Cl(-)/HCO(3)(-) exchange. These include SLC26A3, A4, A6, A7, A9, and A11, which are expressed in the kidney except for SLC26A3 (DRA), which is predominantly expressed in the intestine. SLC26 Cl(-)/HCO(3)(-) exchanger isoforms display unique nephron segment distribution patterns with distinct subcellular localization in the kidney tubules. Together with studies in pathophysiologic states and the examination of genetically engineered mouse models, the evolving picture points to important roles for the SLC26 family in health and disease states. This review summarizes recent advances in the characterization of the SLC26 Cl(-)/HCO(3)(-) exchangers in the kidney with emphasis on their essential role in diverse physiological processes, including chloride homeostasis, oxalate excretion and kidney stone formation, vascular volume and blood pressure regulation, and acid-base balance.
Collapse
Affiliation(s)
- Manoocher Soleimani
- 1] Center on Genetics of Transport and Epithelial Biology, University of Cincinnati, Cincinnati, Ohio, USA [2] Research Services, Veterans Affairs Medical Center, Cincinnati, Ohio, USA [3] Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
46
|
Alper SL, Sharma AK. The SLC26 gene family of anion transporters and channels. Mol Aspects Med 2013; 34:494-515. [PMID: 23506885 DOI: 10.1016/j.mam.2012.07.009] [Citation(s) in RCA: 272] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/21/2012] [Indexed: 02/08/2023]
Abstract
The phylogenetically ancient SLC26 gene family encodes multifunctional anion exchangers and anion channels transporting a broad range of substrates, including Cl(-), HCO3(-), sulfate, oxalate, I(-), and formate. SLC26 polypeptides are characterized by N-terminal cytoplasmic domains, 10-14 hydrophobic transmembrane spans, and C-terminal cytoplasmic STAS domains, and appear to be homo-oligomeric. SLC26-related SulP proteins of marine bacteria likely transport HCO3(-) as part of oceanic carbon fixation. SulP genes present in antibiotic operons may provide sulfate for antibiotic biosynthetic pathways. SLC26-related Sultr proteins transport sulfate in unicellular eukaryotes and in plants. Mutations in three human SLC26 genes are associated with congenital or early onset Mendelian diseases: chondrodysplasias for SLC26A2, chloride diarrhea for SLC26A3, and deafness with enlargement of the vestibular aqueduct for SLC26A4. Additional disease phenotypes evident only in mouse knockout models include oxalate urolithiasis for Slc26a6 and Slc26a1, non-syndromic deafness for Slc26a5, gastric hypochlorhydria for Slc26a7 and Slc26a9, distal renal tubular acidosis for Slc26a7, and male infertility for Slc26a8. STAS domains are required for cell surface expression of SLC26 proteins, and contribute to regulation of the cystic fibrosis transmembrane regulator in complex, cell- and tissue-specific ways. The protein interactomes of SLC26 polypeptides are under active investigation.
Collapse
Affiliation(s)
- Seth L Alper
- Renal Division and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| | | |
Collapse
|
47
|
Wall SM, Weinstein AM. Cortical distal nephron Cl(-) transport in volume homeostasis and blood pressure regulation. Am J Physiol Renal Physiol 2013; 305:F427-38. [PMID: 23637202 DOI: 10.1152/ajprenal.00022.2013] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Renal intercalated cells mediate the secretion or absorption of Cl(-) and OH(-)/H(+) equivalents in the connecting segment (CNT) and cortical collecting duct (CCD). In so doing, they regulate acid-base balance, vascular volume, and blood pressure. Cl(-) absorption is either electrogenic and amiloride-sensitive or electroneutral and thiazide-sensitive. However, which Cl(-) transporter(s) are targeted by these diuretics is debated. While epithelial Na(+) channel (ENaC) does not transport Cl(-), it modulates Cl(-) transport probably by generating a lumen-negative voltage, which drives Cl(-) flux across tight junctions. In addition, recent evidence indicates that ENaC inhibition increases electrogenic Cl(-) secretion via a type A intercalated cells. During ENaC blockade, Cl(-) is taken up across the basolateral membrane through the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1) and then secreted across the apical membrane through a conductive pathway (a Cl(-) channel or an electrogenic exchanger). The mechanism of this apical Cl(-) secretion is unresolved. In contrast, thiazide diuretics inhibit electroneutral Cl(-) absorption mediated by a Na(+)-dependent Cl(-)/HCO3(-) exchanger. The relative contribution of the thiazide and the amiloride-sensitive components of Cl(-) absorption varies between studies and probably depends on the treatment model employed. Cl(-) absorption increases markedly with angiotensin and aldosterone administration, largely by upregulating the Na(+)-independent Cl(-)/HCO3(-) exchanger pendrin. In the absence of pendrin [Slc26a4((-/-)) or pendrin null mice], aldosterone-stimulated Cl(-) absorption is significantly reduced, which attenuates the pressor response to this steroid hormone. Pendrin also modulates aldosterone-induced changes in ENaC abundance and function through a kidney-specific mechanism that does not involve changes in the concentration of a circulating hormone. Instead, pendrin changes ENaC abundance and function, at least in part, by altering luminal HCO3(-). This review summarizes mechanisms of Cl(-) transport in CNT and CCD and how these transporters contribute to the regulation of extracellular volume and blood pressure.
Collapse
Affiliation(s)
- Susan M Wall
- Renal Division, WMB Rm. 338, 1639 Pierce Dr., NE, Atlanta, GA 30322.
| | | |
Collapse
|
48
|
Amlal H, Xu J, Barone S, Zahedi K, Soleimani M. The chloride channel/transporter Slc26a9 regulates the systemic arterial pressure and renal chloride excretion. J Mol Med (Berl) 2013; 91:561-72. [PMID: 23149824 PMCID: PMC11709006 DOI: 10.1007/s00109-012-0973-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 10/07/2012] [Accepted: 10/16/2012] [Indexed: 01/21/2023]
Abstract
Apical chloride secretory pathways in the kidney medullary collecting duct are thought to play an important role in the modulation of final urine composition and regulation of systemic vascular volume and/or blood pressure. However, the molecular identity of these molecules has largely remained unknown. Here, we demonstrate that Slc26a9, an electrogenic chloride channel/transporter, is localized on the apical membrane of principal cells in the kidney medullary collecting duct and mediates chloride secretion. Mice with the genetic deletion of Slc26a9 show significant reduction in renal chloride excretion when fed a diet high in salt or subjected to water deprivation. Arterial pressure measurements indicated that Slc26a9 knockout (Slc26a9(-/-)) mice are hypertensive under baseline conditions and increase their blood pressure further within 48 h of switching to a high-salt diet. These results suggest that Slc26a9 plays an important role in renal chloride/fluid excretion and arterial pressure regulation. We propose that impaired SLC26A9 activity in humans may interfere with the excretion of excess salt and result in hypertension.
Collapse
Affiliation(s)
- Hassane Amlal
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, USA
| | | | | | | | | |
Collapse
|
49
|
Li HC, Du Z, Barone S, Rubera I, McDonough AA, Tauc M, Zahedi K, Wang T, Soleimani M. Proximal tubule specific knockout of the Na⁺/H⁺ exchanger NHE3: effects on bicarbonate absorption and ammonium excretion. J Mol Med (Berl) 2013; 91:951-63. [PMID: 23508938 PMCID: PMC3730089 DOI: 10.1007/s00109-013-1015-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/16/2013] [Accepted: 02/25/2013] [Indexed: 11/17/2022]
Abstract
The existing NHE3 knockout mouse has significant intestinal electrolyte absorption defects, making this model unsuitable for the examination of the role of proximal tubule NHE3 in pathophysiologic states in vivo. To overcome this problem, we generated proximal convoluted tubule-specific KO mice (NHE3-PT KO) by generating and crossing NHE3 floxed mice with the sodium-glucose transporter 2 Cre transgenic mice. The NHE3-PT KO mice have >80 % ablation of NHE3 as determined by immunofluorescence microscopy, western blot, and northern analyses, and show mild metabolic acidosis (serum bicarbonate of 21.2 mEq/l in KO vs. 23.7 mEq/l in WT, p < 0.05). In vitro microperfusion studies in the isolated proximal convoluted tubules demonstrated a ∼36 % reduction in bicarbonate reabsorption (JHCO3 = 53.52 ± 4.61 pmol/min/mm in KO vs. 83.09 ± 9.73 in WT) and a ∼27 % reduction in volume reabsorption (Jv = 0.67 ± 0.07 nl/min/mm in KO vs. 0.92 ± 0.06 nl/min/mm in WT) in mutant mice. The NHE3-PT KO mice tolerated NH4Cl acid load well (added to the drinking water) and showed NH4 excretion rates comparable to WT mice at 2 and 5 days after NH4Cl loading without disproportionate metabolic acidosis after 5 days of acid load. Our results suggest that the Na+/H+ exchanger NHE3 plays an important role in fluid and bicarbonate reabsorption in the proximal convoluted tubule but does not play an important role in NH4 excretion.
Collapse
Affiliation(s)
- Hong C Li
- Center on Genetics of Transport and the Department of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Song Y, Yamamoto A, Steward MC, Ko SBH, Stewart AK, Soleimani M, Liu BC, Kondo T, Jin CX, Ishiguro H. Deletion of Slc26a6 alters the stoichiometry of apical Cl-/HCO-3 exchange in mouse pancreatic duct. Am J Physiol Cell Physiol 2012; 303:C815-24. [PMID: 22895259 DOI: 10.1152/ajpcell.00151.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
To define the stoichiometry and molecular identity of the Cl(-)/HCO(3)(-) exchanger in the apical membrane of pancreatic duct cells, changes in luminal pH and volume were measured simultaneously in interlobular pancreatic ducts isolated from wild-type and Slc26a6-null mice. Transepithelial fluxes of HCO(3)(-) and Cl(-) were measured in the presence of anion gradients favoring rapid exchange of intracellular HCO(3)(-) with luminal Cl(-) in cAMP-stimulated ducts. The flux ratio of Cl(-) absorption/HCO(3)(-) secretion was ∼0.7 in wild-type ducts and ∼1.4 in Slc26a6(-/-) ducts where a different Cl(-)/HCO(3)(-) exchanger, most likely SLC26A3, was found to be active. Interactions between Cl(-)/HCO(3)(-) exchange and cystic fibrosis transmembrane conductance regulator (CFTR) in cAMP-stimulated ducts were examined by measuring the recovery of intracellular pH after alkali-loading by acetate prepulse. Hyperpolarization induced by luminal application of CFTRinh-172 enhanced HCO(3)(-) efflux across the apical membrane via SLC26A6 in wild-type ducts but significantly reduced HCO(3)(-) efflux in Slc26a6(-/-) ducts. In microperfused wild-type ducts, removal of luminal Cl(-), or luminal application of dihydro-4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid to inhibit SLC26A6, caused membrane hyperpolarization, which was abolished in Slc26a6(-/-) ducts. In conclusion, we have demonstrated that deletion of Slc26a6 alters the apparent stoichiometry of apical Cl(-)/HCO(3)(-) exchange in native pancreatic duct. Our results are consistent with SLC26A6 mediating 1:2 Cl(-)/HCO(3)(-) exchange, and the exchanger upregulated in its absence, most probably SLC26A3, mediating 2:1 exchange.
Collapse
Affiliation(s)
- Ying Song
- Laboratory of Human Nutrition, Nagoya University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|