1
|
Lee HJ, Sun Y, Das F, Ju W, Nair V, Kevil CG, Varadarajan S, Zhang G, Ghosh Choudhury G, Singh BB, Kretzler M, Nelson RG, Sharma K, Kasinath BS. The CLCA1/TMEM16A/Cl- current axis associates with H2S deficiency in diabetic kidney injury. JCI Insight 2025; 10:e174848. [PMID: 39782685 PMCID: PMC11721299 DOI: 10.1172/jci.insight.174848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/13/2024] [Indexed: 01/30/2025] Open
Abstract
The role played by anionic channels in diabetic kidney disease (DKD) is not known. Chloride channel accessory 1 (CLCA1) facilitates the activity of TMEM16A (Anoctamin-1), a Ca2+-dependent Cl- channel. We examined if CLCA1/TMEM16A had a role in DKD. In mice with type 2 diabetes, renal cortical CLCA1 and TMEM16A content was increased. CLCA1 and TMEM16A content was associated with hydrogen sulfide (H2S) deficiency, mTOR complex 1 (mTORC1) activation, albuminuria, and matrix increase. Administering sodium hydrosulfide (NaHS), a source of H2S, mitigated these changes. In proximal tubular epithelial (MCT) cells, high glucose rapidly increased CLCA1 by recruiting the IL-6/STAT3 axis and augmented TMEM16A expression by stimulating its mRNA translation; these changes were abolished by NaHS. Patch clamp experiments showed that high glucose increased Cl- current in MCT cells that was ameliorated by NaHS and a TMEM16A chemical inhibitor. siRNA against CLCA1 or TMEM16A and TMEM16A inhibitor abolished high glucose-induced mTORC1 activation and matrix protein increase. Tubular expression of TMEM16A correlated with albuminuria in kidney biopsies from people with type 2 diabetes. We report a pathway for DKD in which H2S deficiency results in kidney injury by the recruitment of the CLCA1/TMEM16A/Cl- current system.
Collapse
Affiliation(s)
- Hak Joo Lee
- Center for Precision Medicine, Department of Medicine, and
| | - Yuyang Sun
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Falguni Das
- Center for Precision Medicine, Department of Medicine, and
| | - Wenjun Ju
- Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Christopher G. Kevil
- Department of Pathology, Louisiana State University Health Science Center, Shreveport, Louisiana, USA
| | - Shankara Varadarajan
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Guanshi Zhang
- Center for Precision Medicine, Department of Medicine, and
| | - Goutam Ghosh Choudhury
- Center for Precision Medicine, Department of Medicine, and
- Research service and
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | - Brij B. Singh
- Department of Periodontics, University of Texas Health, San Antonio, San Antonio, Texas, USA
| | - Matthias Kretzler
- Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Robert G. Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
- Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Kumar Sharma
- Center for Precision Medicine, Department of Medicine, and
- Research service and
| | - Balakuntalam S. Kasinath
- Center for Precision Medicine, Department of Medicine, and
- Research service and
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
2
|
Chen R, Zou J, Chen J, Wang L, Kang R, Tang D. Immune aging and infectious diseases. Chin Med J (Engl) 2024; 137:3010-3049. [PMID: 39679477 PMCID: PMC11706578 DOI: 10.1097/cm9.0000000000003410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT The rise in global life expectancy has led to an increase in the older population, presenting significant challenges in managing infectious diseases. Aging affects the innate and adaptive immune systems, resulting in chronic low-grade inflammation (inflammaging) and immune function decline (immunosenescence). These changes would impair defense mechanisms, increase susceptibility to infections and reduce vaccine efficacy in older adults. Cellular senescence exacerbates these issues by releasing pro-inflammatory factors, further perpetuating chronic inflammation. Moreover, comorbidities, such as cardiovascular disease and diabetes, which are common in older adults, amplify immune dysfunction, while immunosuppressive medications further complicate responses to infections. This review explores the molecular and cellular mechanisms driving inflammaging and immunosenescence, focusing on genomic instability, telomere attrition, and mitochondrial dysfunction. Additionally, we discussed how aging-associated immune alterations influence responses to bacterial, viral, and parasitic infections and evaluated emerging antiaging strategies, aimed at mitigating these effects to improve health outcomes in the aging population.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ju Zou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Jiawang Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ling Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
3
|
Lee HJ, Min L, Gao J, Matta S, Drel V, Saliba A, Tamayo I, Montellano R, Hejazi L, Maity S, Xu G, Grajeda BI, Roy S, Hallows KR, Choudhury GG, Kasinath BS, Sharma K. Female Protection Against Diabetic Kidney Disease Is Regulated by Kidney-Specific AMPK Activity. Diabetes 2024; 73:1167-1177. [PMID: 38656940 PMCID: PMC11189830 DOI: 10.2337/db23-0807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
Reduced kidney AMPK activity is associated with nutrient stress-induced chronic kidney disease (CKD) in male mice. In contrast, female mice resist nutrient stress-induced CKD. The role of kidney AMPK in sex-related organ protection against nutrient stress and metabolite changes was evaluated in diabetic kidney tubule-specific AMPKγ2KO (KTAMPKγ2ΚΟ) male and female mice. In wild-type (WT) males, diabetes increased albuminuria, urinary kidney injury molecule-1, hypertension, kidney p70S6K phosphorylation, and kidney matrix accumulation; these features were not exacerbated with KTAMPKγ2ΚΟ. Whereas WT females had protection against diabetes-induced kidney injury, KTAMPKγ2ΚΟ led to loss of female protection against kidney disease. The hormone 17β-estradiol ameliorated high glucose-induced AMPK inactivation, p70S6K phosphorylation, and matrix protein accumulation in kidney tubule cells. The mechanism for female protection against diabetes-induced kidney injury is likely via an estrogen-AMPK pathway, as inhibition of AMPK led to loss of estrogen protection to glucose-induced mTORC1 activation and matrix production. RNA sequencing and metabolomic analysis identified a decrease in the degradation pathway of phenylalanine and tyrosine resulting in increased urinary phenylalanine and tyrosine levels in females. The metabolite levels correlated with loss of female protection. The findings provide new insights to explain evolutionary advantages to females during states of nutrient challenges. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Hak Joo Lee
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| | - Liang Min
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Jingli Gao
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Shane Matta
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Viktor Drel
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Afaf Saliba
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Ian Tamayo
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Richard Montellano
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Leila Hejazi
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Soumya Maity
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Guogang Xu
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
| | - Brian I. Grajeda
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas, El Paso, TX
| | - Sourav Roy
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas, El Paso, TX
| | - Kenneth R. Hallows
- USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Goutam Ghosh Choudhury
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| | - Balakuntalam S. Kasinath
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| | - Kumar Sharma
- Center for Precision Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX
- South Texas Veterans Health Care System, San Antonio, TX
| |
Collapse
|
4
|
Sala-Rabanal M, Yurtsever Z, Berry KN, McClenaghan C, Foy AJ, Hanson A, Steinberg DF, Greven JA, Kluender CE, Alexander-Brett JM, Nichols CG, Brett TJ. Modulation of TMEM16B channel activity by the calcium-activated chloride channel regulator 4 (CLCA4) in human cells. J Biol Chem 2024; 300:107432. [PMID: 38825009 PMCID: PMC11231702 DOI: 10.1016/j.jbc.2024.107432] [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: 04/08/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024] Open
Abstract
The Ca2+-activated Cl- channel regulator CLCA1 potentiates the activity of the Ca2+-activated Cl- channel (CaCC) TMEM16A by directly engaging the channel at the cell surface, inhibiting its reinternalization and increasing Ca2+-dependent Cl- current (ICaCC) density. We now present evidence of functional pairing between two other CLCA and TMEM16 protein family members, namely CLCA4 and the CaCC TMEM16B. Similar to CLCA1, (i) CLCA4 is a self-cleaving metalloprotease, and the N-terminal portion (N-CLCA4) is secreted; (ii) the von Willebrand factor type A (VWA) domain in N-CLCA4 is sufficient to potentiate ICaCC in HEK293T cells; and (iii) this is mediated by the metal ion-dependent adhesion site motif within VWA. The results indicate that, despite the conserved regulatory mechanism and homology between CLCA1 and CLCA4, CLCA4-dependent ICaCC are carried by TMEM16B, rather than TMEM16A. Our findings show specificity in CLCA/TMEM16 interactions and suggest broad physiological and pathophysiological links between these two protein families.
Collapse
Affiliation(s)
- Monica Sala-Rabanal
- Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Zeynep Yurtsever
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Kayla N Berry
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA; Immunology Program and Medical Scientist Training Program, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Alyssa J Foy
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Alex Hanson
- Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Deborah F Steinberg
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jessica A Greven
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Colin E Kluender
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jennifer M Alexander-Brett
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Tom J Brett
- Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri, USA; Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
5
|
Morse J, Wang D, Mei S, Whitham D, Hladun C, Darie CC, Sintim HO, Wang M, Leung K. Chloride Homeostasis Regulates cGAS-STING Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588475. [PMID: 38645072 PMCID: PMC11030317 DOI: 10.1101/2024.04.08.588475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The cGAS-STING signaling pathway has emerged as a key mediator of inflammation. However, the roles of chloride homeostasis on this pathway are unclear. Here, we uncovered a correlation between chloride homeostasis and cGAS-STING signaling. We found that dysregulation of chloride homeostasis attenuates cGAS-STING signaling in a lysosome-independent manner. Treating immune cells with chloride channel inhibitors attenuated 2'3'-cGAMP production by cGAS and also suppressed STING polymerization, leading to reduced cytokine production. We also demonstrate that non-selective chloride channel blockers can suppress the NPC1 deficiency-induced, hyper-activated STING signaling in skin fibroblasts derived from Niemann Pick disease type C (NPC) patients. Our findings reveal that chloride homeostasis majorly affects cGAS-STING pathway and suggest a provocative strategy to dampen STING-mediated inflammation via targeting chloride channels.
Collapse
Affiliation(s)
- Jared Morse
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danna Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Serena Mei
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Danielle Whitham
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Colby Hladun
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Costel C. Darie
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - Herman O. Sintim
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Modi Wang
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| | - KaHo Leung
- Department of Chemistry & Biomolecular Science, Clarkson University, NY, 13676, United States
| |
Collapse
|
6
|
Schaub JA, O'Connor CL, Dailey M, Hlynka AW, Chang Y, Postiff D, Kaffenberger SD, Palapattu GS, Gillespie BW, Hodgin JB, Shedden K, Bitzer M. Spatial Heterogeneity of Glomerular Phenotypes Affects Kidney Biopsy Findings. KIDNEY360 2023; 4:1598-1607. [PMID: 37889598 PMCID: PMC10695647 DOI: 10.34067/kid.0000000000000283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023]
Abstract
Key Points Glomeruli with pathologic changes are not homogeneously distributed throughout the kidney cortex. Biopsies that do not include the kidney capsule may underdetect glomeruli with pathologic changes. Location of glomeruli with pathologic changes may be related to underlying clinical characteristics. Background Detection of rare glomerular phenotypes can affect diagnosis in indication kidney biopsies and in kidney tissue used for research studies. Nephropathologists are aware of potential sampling error when assessing needle biopsy cores, but quantitative data are lacking. Methods Kidney tissue from patients undergoing total nephrectomy enrolled in an observational, cross-sectional cohort study was used to characterize glomeruli as typical or atypical, which included globally sclerotic glomeruli (GSGs), segmentally sclerotic glomeruli, ischemic-like, and imploding. A 2D map of the glomerular annotations was generated. Spatial centrality of atypical glomeruli using the L2 metric and differences in pairwise distances between typical or atypical glomeruli were calculated. To determine how the yield of capturing atypical glomerular phenotype was affected by biopsy depth (i.e. , not including the renal capsule), simulated kidney biopsies were generated from the 2D map. Results The mean number of glomeruli in a nephrectomy specimen was 209 (SD 143), and GSGs were the most common type of atypical glomeruli (median: 13% [interquartile range: 5,31]). Typical glomeruli were more likely to be surrounded by other glomeruli (i.e. , centrally located in the kidney cortex) than GSGs, segmentally sclerosed glomeruli, ischemic-like glomeruli, and imploding glomeruli. Atypical glomeruli were 7.3% (95% confidence interval, 4.1 to 10.4) closer together than typical glomeruli and were more likely to be closer together in older patients or those with hypertension. In simulated kidney biopsies, failure to capture the capsule was associated with underdetection of GSGs, ischemic-like glomeruli, and imploding glomeruli. Conclusions Spatial analysis of large sections of kidney tissue provided quantitative evidence of spatial heterogeneity of glomerular phenotypes including clustering of atypical glomeruli in individuals with hypertension or older age. Most importantly, deep kidney biopsies that lack subcapsular area underdetect atypical glomerular phenotypes, suggesting that capturing the renal capsule is an important quality control measure for kidney biopsies.
Collapse
Affiliation(s)
- Jennifer A. Schaub
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | - Meghan Dailey
- Advanced Research Computing (Information and Technology Services), University of Michigan, Ann Arbor, Michigan
| | | | - Yurui Chang
- Department of Statistics, University of Michigan, Ann Arbor, Michigan
| | - Deborah Postiff
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | | | - Brenda W. Gillespie
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Jeffrey B. Hodgin
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Kerby Shedden
- Department of Statistics, University of Michigan, Ann Arbor, Michigan
| | - Markus Bitzer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
7
|
Zhang Y, Xie Y, Lu W, Xu S, Wang X, Zhou W, Zhang Y, Ding X, Zhao S. Identification of resident progenitors labeled with Top2a responsible for proximal tubular regeneration in ischemia reperfusion-induced acute kidney injury. Cell Signal 2023; 101:110506. [PMID: 36309330 DOI: 10.1016/j.cellsig.2022.110506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Acute kidney injury is a common fatal disease with complex etiology and limited treatment methods. Proximal tubules (PTs) are the most vulnerable segment. Not only in injured kidneys but also in normal kidneys, shedding of PTs often happens. However, the source cells and mechanism of their regeneration remain unclear. METHODS ScRNA and snRNA sequencing data of acute injured or normal kidney were downloaded from GEO database to identify the candidate biomarker of progenitor of proximal tubules. SLICE algorithm and CytoTRACE analyses were employed to evaluate the stemness of progenitors. Then the repairing trajectory was constructed through pseudotime analyses. SCENIC algorithm was used to detect cell-type-specific regulon. With spatial transcriptome data, the location of progenitors was simulated. Neonatal/ adult/ aged mice and preconditioning AKI mice model and deconvolution of 2 RNA-seq data were employed for validation. RESULTS Through cluster identification, PT cluster expressed Top2a specifically was identified to increase significantly during AKI. With relatively strong stemness, the Top2a-labeled PT cluster tended to be the origin of the repairing trajectory. Moreover, the cluster was regulated by Pbx3-based regulon and possessed great segmental heterogeneity. Changes of Top2a between neonatal and aged mice and among AKI models validated the progenitor role of Top2a-labeled cluster. CONCLUSIONS Our study provided transcriptomic evidence that resident proximal tubular progenitors labeled with Top2a participated in regeneration. Considering the segmental heterogeneity, we find that there is a group of reserve progenitor cells in each tubular segment. When AKI occurs, the reserve progenitors of each tubular segment proliferate and replenish first, and PT-progenitors, a cluster with no obvious PT markers replenish each subpopulation of the reserve cells.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Yeqing Xie
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney Disease; Kidney and Dialysis Institute of Shanghai; Kidney and Blood Purification Key Laboratory of Shanghai
| | - Wei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Sujuan Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Xiaoyan Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Weiran Zhou
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Yingjia Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney Disease; Kidney and Dialysis Institute of Shanghai; Kidney and Blood Purification Key Laboratory of Shanghai.
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney Disease; Kidney and Dialysis Institute of Shanghai; Kidney and Blood Purification Key Laboratory of Shanghai.
| |
Collapse
|
8
|
Chen R, Cao C, Liu H, Jiang W, Pan R, He H, Ding K, Meng Q. Macrophage Sprouty4 deficiency diminishes sepsis-induced acute lung injury in mice. Redox Biol 2022; 58:102513. [PMID: 36334381 PMCID: PMC9637958 DOI: 10.1016/j.redox.2022.102513] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/10/2022] [Accepted: 10/15/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Inflammation and oxidative stress play critical roles in sepsis-induced acute lung injury (ALI). Sprout4 (Spry4) is involved in regulating inflammation and tissue injury; however, its role and mechanism in sepsis-induced ALI remain elusive. METHODS Macrophage-specific Spry4 knockout (Spry4MKO), transgenic (Spry4MTG) mice and matched control littermates were generated and exposed to cecum ligation and puncture (CLP) surgery to establish bacterial sepsis-induced ALI. Bone marrow-derived macrophages (BMDMs) from Spry4MKO or Spry4MTG mice were isolated and subjected to lipopolysaccharide (LPS) stimulation to further validate the role of Spry4 in vitro. To verify the necessity of AMP-activated protein kinase (AMPK), Spry4 and AMPK double knockout mice and compound C were used in vivo and in vitro. BMDMs were treated with STO-609 to inhibit calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2). RESULTS We found that macrophage Spry4 was increased in CLP mice and positively correlated with sepsis-induced ALI. Macrophage Spry4 deficiency prevented, while macrophage Spry4 overexpression exacerbated sepsis-induced inflammation, oxidative stress and ALI in mice and BMDMs. Mechanistic studies revealed that macrophage Spry4 deficiency alleviated sepsis-induced ALI through activating CaMKK2/AMPK pathway. CONCLUSION Our study identify macrophage Spry4 as a promising predictive and therapeutic target of sepsis-induced ALI.
Collapse
Affiliation(s)
- Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chen Cao
- Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Huimin Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Pan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - He He
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qingtao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
9
|
Yan P, Ke B, Fang X. Ion channels as a therapeutic target for renal fibrosis. Front Physiol 2022; 13:1019028. [PMID: 36277193 PMCID: PMC9581181 DOI: 10.3389/fphys.2022.1019028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Renal ion channel transport and electrolyte disturbances play an important role in the process of functional impairment and fibrosis in the kidney. It is well known that there are limited effective drugs for the treatment of renal fibrosis, and since a large number of ion channels are involved in the renal fibrosis process, understanding the mechanisms of ion channel transport and the complex network of signaling cascades between them is essential to identify potential therapeutic approaches to slow down renal fibrosis. This review summarizes the current work of ion channels in renal fibrosis. We pay close attention to the effect of cystic fibrosis transmembrane conductance regulator (CFTR), transmembrane Member 16A (TMEM16A) and other Cl− channel mediated signaling pathways and ion concentrations on fibrosis, as well as the various complex mechanisms for the action of Ca2+ handling channels including Ca2+-release-activated Ca2+ channel (CRAC), purinergic receptor, and transient receptor potential (TRP) channels. Furthermore, we also focus on the contribution of Na+ transport such as epithelial sodium channel (ENaC), Na+, K+-ATPase, Na+-H+ exchangers, and K+ channels like Ca2+-activated K+ channels, voltage-dependent K+ channel, ATP-sensitive K+ channels on renal fibrosis. Proposed potential therapeutic approaches through further dissection of these mechanisms may provide new therapeutic opportunities to reduce the burden of chronic kidney disease.
Collapse
|
10
|
Pippin JW, Kaverina N, Wang Y, Eng DG, Zeng Y, Tran U, Loretz CJ, Chang A, Akilesh S, Poudel C, Perry HS, O’Connor C, Vaughan JC, Bitzer M, Wessely O, Shankland SJ. Upregulated PD-1 signaling antagonizes glomerular health in aged kidneys and disease. J Clin Invest 2022; 132:e156250. [PMID: 35968783 PMCID: PMC9374384 DOI: 10.1172/jci156250] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/24/2022] [Indexed: 01/21/2023] Open
Abstract
With an aging population, kidney health becomes an important medical and socioeconomic factor. Kidney aging mechanisms are not well understood. We previously showed that podocytes isolated from aged mice exhibit increased expression of programmed cell death protein 1 (PD-1) surface receptor and its 2 ligands (PD-L1 and PD-L2). PDCD1 transcript increased with age in microdissected human glomeruli, which correlated with lower estimated glomerular filtration rate and higher segmental glomerulosclerosis and vascular arterial intima-to-lumen ratio. In vitro studies in podocytes demonstrated a critical role for PD-1 signaling in cell survival and in the induction of a senescence-associated secretory phenotype. To prove PD-1 signaling was critical to podocyte aging, aged mice were injected with anti-PD-1 antibody. Treatment significantly improved the aging phenotype in both kidney and liver. In the glomerulus, it increased the life span of podocytes, but not that of parietal epithelial, mesangial, or endothelial cells. Transcriptomic and immunohistochemistry studies demonstrated that anti-PD-1 antibody treatment improved the health span of podocytes. Administering the same anti-PD-1 antibody to young mice with experimental focal segmental glomerulosclerosis (FSGS) lowered proteinuria and improved podocyte number. These results suggest a critical contribution of increased PD-1 signaling toward both kidney and liver aging and in FSGS.
Collapse
Affiliation(s)
| | | | - Yuliang Wang
- Paul G. Allen School of Computer Science and Engineering, and
| | | | - Yuting Zeng
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Uyen Tran
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | | | - Anthony Chang
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Shreeram Akilesh
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Hannah S. Perry
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | | | - Joshua C. Vaughan
- Department of Chemistry, University of Washington, Seattle, Washington, USA
- Department of Physiology and Biophysics and
| | - Markus Bitzer
- Division of Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Oliver Wessely
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Stuart J. Shankland
- Division of Nephrology
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Tan H, Xu J, Liu Y. Ageing, cellular senescence and chronic kidney disease: experimental evidence. Curr Opin Nephrol Hypertens 2022; 31:235-243. [PMID: 35142744 PMCID: PMC9035037 DOI: 10.1097/mnh.0000000000000782] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) is often viewed as an accelerated and premature ageing of the kidney, as they share common pathological features characterized by cellular senescence. In this review, we summarize the experimental evidence linking cellular senescence to the pathobiology of kidney ageing and CKD, and discuss the strategies for targeting senescent cells in developing therapeutics for ageing-related kidney disorders. RECENT FINDINGS Kidney ageing and CKD are featured with increased cellular senescence, an irreversible state of cell cycle arrest and the cessation of cell division. Senescent cells secrete a diverse array of proinflammatory and profibrotic factors known as senescence-associated secretory phenotype (SASP). Secondary senescence can be induced by primary senescent cells via a mechanism involving direct contact or the SASP. Various senolytic therapies aiming to selectively remove senescent cells in vivo have been developed. Senostatic approaches to suppress senescence or inhibit SASP, as well as nutrient signalling regulators are also validated in animal models of ageing. SUMMARY These recent studies provide experimental evidence supporting the notion that accumulation of senescent cells and their associated SASP is a main driver leading to structural and functional organ degeneration in CKD and other ageing-related disorder.
Collapse
Affiliation(s)
- Huishi Tan
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Lee HJ, Donati A, Feliers D, Sun Y, Ding Y, Madesh M, Salmon AB, Ikeno Y, Ross C, O'Connor CL, Ju W, Bitzer M, Chen Y, Choudhury GG, Singh BB, Sharma K, Kasinath BS. Chloride channel accessory 1 integrates chloride channel activity and mTORC1 in aging-related kidney injury. Aging Cell 2021; 20:e13407. [PMID: 34118180 PMCID: PMC8282273 DOI: 10.1111/acel.13407] [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] [Received: 10/27/2020] [Revised: 03/30/2021] [Accepted: 05/08/2021] [Indexed: 12/19/2022] Open
Abstract
The mechanism of kidney injury in aging are not well understood. In order to identify hitherto unknown pathways of aging‐related kidney injury, we performed RNA‐Seq on kidney extracts of young and aged mice. Expression of chloride (Cl) channel accessory 1 (CLCA1) mRNA and protein was increased in the kidneys of aged mice. Immunostaining showed a marked increase in CLCLA1 expression in the proximal tubules of the kidney from aged mice. Increased kidney CLCA1 gene expression also correlated with aging in marmosets and in a human cohort. In aging mice, increased renal cortical CLCA1 content was associated with hydrogen sulfide (H2S) deficiency, which was ameliorated by administering sodium hydrosulfide (NaHS), a source of H2S. In order to study whether increased CLCA1 expression leads to injury phenotype and the mechanisms involved, stable transfection of proximal tubule epithelial cells overexpressing human CLCA1 (hCLCA1) was performed. Overexpression of hCLCA1 augmented Cl− current via the Ca++‐dependent Cl− channel TMEM16A (anoctamin‐1) by patch‐clamp studies. hCLCA1 overexpression also increased the expression of fibronectin, a matrix protein, and induced the senescence‐associated secretory phenotype (SASP). Mechanistic studies underlying these changes showed that hCLCA1 overexpression leads to inhibition of AMPK activity and stimulation of mTORC1 as cellular signaling determinants of injury. Both TMEM16A inhibitor and NaHS reversed these signaling events and prevented changes in fibronectin and SASP. We conclude that CLCA1‐TMEM16A‐Cl− current pathway is a novel mediator of kidney injury in aging that is regulated by endogenous H2S.
Collapse
Affiliation(s)
- Hak Joo Lee
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Andrew Donati
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Denis Feliers
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Yuyang Sun
- Department of Periodontics University of Texas Health San Antonio TX USA
| | - Yanli Ding
- Department of Pathology University of Texas Health San Antonio TX USA
| | - Muniswamy Madesh
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Adam B. Salmon
- Department of Molecular Medicine University of Texas Health San Antonio TX USA
- Barshop Institute for Longevity and Aging Studies University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
- Geriatric Research Education & Clinical Center South Texas Veterans Health Care System San Antonio TX USA
| | - Yuji Ikeno
- Department of Pathology University of Texas Health San Antonio TX USA
- Department of Molecular Medicine University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
| | - Corinna Ross
- Texas Biomedical Research Institute Southwest National Primate Research Center San Antonio TX USA
- Department of Science and Mathematics Texas A&M University San Antonio San Antonio TX USA
| | | | - Wenjun Ju
- Department of Internal Medicine University of Michigan Ann Arbor MI USA
| | - Markus Bitzer
- Department of Internal Medicine University of Michigan Ann Arbor MI USA
| | - Yidong Chen
- Department of Population Health Sciences University of Texas Health San Antonio TX USA
- Greehey Children's Cancer Research Institute University of Texas Health San Antonio TX USA
| | - Goutam Ghosh Choudhury
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
- Geriatric Research Education & Clinical Center South Texas Veterans Health Care System San Antonio TX USA
| | - Brij B. Singh
- Department of Periodontics University of Texas Health San Antonio TX USA
| | - Kumar Sharma
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
| | - Balakuntalam S. Kasinath
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
- Barshop Institute for Longevity and Aging Studies University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
- Geriatric Research Education & Clinical Center South Texas Veterans Health Care System San Antonio TX USA
| |
Collapse
|