1
|
Pernecker M, Ciarimboli G. Regulation of renal organic cation transporters. FEBS Lett 2024; 598:2328-2347. [PMID: 38831380 DOI: 10.1002/1873-3468.14943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Transporters for organic cations (OCs) facilitate exchange of positively charged molecules through the plasma membrane. Substrates for these transporters encompass neurotransmitters, metabolic byproducts, drugs, and xenobiotics. Consequently, these transporters actively contribute to the regulation of neurotransmission, cellular penetration and elimination process for metabolic products, drugs, and xenobiotics. Therefore, these transporters have significant physiological, pharmacological, and toxicological implications. In cells of renal proximal tubules, the vectorial secretion pathways for OCs involve expression of organic cation transporters (OCTs) and multidrug and toxin extrusion proteins (MATEs) on basolateral and apical membrane domains, respectively. This review provides an overview of documented regulatory mechanisms governing OCTs and MATEs. Additionally, regulation of these transporters under various pathological conditions is summarized. The expression and functionality of OCTs and MATEs are subject to diverse pre- and post-translational modifications, providing insights into their regulation in various pathological conditions. Typically, in diseases, downregulation of transporter expression is observed, probably as a protective mechanism to prevent additional damage to kidney tissue. This regulation may be attributed to the intricate network of modifications these transporters undergo, shedding light on their dynamic responses in pathological contexts.
Collapse
Affiliation(s)
- Moritz Pernecker
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| |
Collapse
|
2
|
Deng W, Wei X, Dong Z, Zhang J, Huang Z, Na N. Identification of fibroblast activation-related genes in two acute kidney injury models. PeerJ 2021; 9:e10926. [PMID: 33777519 PMCID: PMC7982076 DOI: 10.7717/peerj.10926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/20/2021] [Indexed: 12/27/2022] Open
Abstract
Background Ischemia-reperfusion injury and drug-induced nephrotoxicity are the two most common reasons for acute kidney injury (AKI). However, little attention has been paid to early activation of fibroblasts in the progression of AKI to chronic kidney disease (CKD). The present study aimed to identify related genes and pathways on fibroblast activation in two mouse models of AKI: ischemia-reperfusion injury (IRI) model and folic acid (FA)-induced injury model. Methods The microarray expression profiles of GSE62732 and GSE121190 were downloaded from the GEO database, and the differentially expressed genes (DEGs) was analyzed using the Limma package of R software. Principal component analysis (PCA) was also performed using R. The functional information of gene products was annotated by Gene Ontology (GO) and DAVID online database, and the pathway analysis was carried out by using the Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) database. Protein-protein interactions (PPI) network was constructed by STRING and Cytoscape. Furthermore, in the Hypoxia/Reoxygenation (H/R) model, the morphological changes of cells were observed under microscope and the expression of the hub genes in NRK-49F cells were validated by qRT-PCR assays. Results A total of 457 DEGs were identified. Among these, 215 DEGs were upregulated and 242 DEGs were downregulated in the acute injured samples compared with uninjured samples. The GO enrichment analysis indicated that these DEGs were mainly involved in transport, the oxidation-reduction process, the metabolic process, metal ion binding, hydrolase activity, and oxidoreductase activity. The KEGG analysis revealed that these DEGs were significantly enriched in the PI3K-Akt signaling pathway, protein digestion and absorption pathway, and focal adhesion pathway. The hub genes including Hnf4α, Pck1 and Timp1 were validated by the qRT-PCR assay in NRK-49F cells in the H/R model. Conclusions Hnf4α, Pck1 and Timp-1 may play a pivotal role in the early activation of fibroblasts, providing novel therapeutic strategies for early prediction and treatment of renal fibrosis.
Collapse
Affiliation(s)
- Weiming Deng
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiangling Wei
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhanwen Dong
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinhua Zhang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhengyu Huang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
3
|
Batista LFS, Torrecilha RBP, Silva RB, Utsunomiya YT, Silva TBF, Tomokane TY, Pacheco AD, Bosco AM, Paulan SC, Rossi CN, Costa GNO, Marcondes M, Ciarlini PC, Nunes CM, Matta VLR, Laurenti MD. Chromosomal segments may explain the antibody response cooperation for canine leishmaniasis pathogenesis. Vet Parasitol 2020; 288:109276. [PMID: 33152678 DOI: 10.1016/j.vetpar.2020.109276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/22/2022]
Abstract
Visceral leishmaniasis (VL) is marked by hyperactivation of a humoral response secreting high quantity of immunoglobulins (Igs) that are inaccessible to intracellular parasites. Here we investigated the contributions of the antibody response to the canine leishmaniasis pathogenesis. Using correlation and genome-wide association analysis, we investigated the relationship of anti-Leishmania infantum immunoglobulin classes levels with parasite burden, clinical response, renal/hepatic biochemical, and oxidative stress markers in dogs from endemic areas of VL. Immunoglobulin G (IgG) and IgA were positively correlated with parasite burden on lymph node and blood. Increased IgG, IgA and IgE levels were associated with severe canine leishmaniasis (CanL) whereas IgM was elevated in uninfected exposed dogs. Correlations of IgM, IgG and IgA with creatinine, urea, AST and ALT levels in the serum were suggested an involvement of those Igs with renal and hepatic changes. The correlogram of oxidative radicals and antioxidants revealed a likely relationship of IgM, IgG and IgA with oxidative stress and lipid peroxidation in the blood, suggested as mechanisms mediating tissue damage and CanL worsening. The gene mapping on chromosomal segments associated with the quantitative variation of immunoglobulin classes identified genetic signatures involved with reactive oxygen species generation, phagolysosome maturation and rupture, free iron availability, Th1/Th2 differenciation and, immunoglobulin clearance. The findings demonstrated the roles of the antibody response as resistance or susceptibility markers and mediators of CanL pathogenesis. In addition we pinpointed candidate genes as potential targets for the therapy against the damage caused by exacerbated antibody response and parasitism in VL.
Collapse
Affiliation(s)
- Luís F S Batista
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Rafaela B P Torrecilha
- Departamento De Medicina Veterinária Preventiva e Reprodução Animal, Faculdade De Ciências Agrárias e Veterinárias, Univ Estadual Paulista, Jaboticabal, São Paulo, CEP: 14884-900, Brazil.
| | - Rafaela B Silva
- Escola de Saúde, Universidade Salvador, Salvador, Bahia, CEP: 41720-200, Brazil.
| | - Yuri T Utsunomiya
- Departamento de Apoio, Produção e Saúde Animal, Faculdade de Medicina Veterinária de Araçatuba, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Thaís B F Silva
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Thaíse Y Tomokane
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Acácio D Pacheco
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Anelise M Bosco
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Silvana C Paulan
- Departamento de Apoio, Produção e Saúde Animal, Faculdade de Medicina Veterinária de Araçatuba, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Claudio N Rossi
- Departamento de Clínica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, CEP 05508-270, Brazil.
| | - Gustavo N O Costa
- Departamento De Medicina Veterinária Preventiva e Reprodução Animal, Faculdade De Ciências Agrárias e Veterinárias, Univ Estadual Paulista, Jaboticabal, São Paulo, CEP: 14884-900, Brazil.
| | - Mary Marcondes
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Paulo C Ciarlini
- Departamento de Clínica, Cirurgia e Reprodução Animal, Faculdade de Medicina Veterinária, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Cáris M Nunes
- Departamento de Apoio, Produção e Saúde Animal, Faculdade de Medicina Veterinária de Araçatuba, Univ Estadual Paulista, Araçatuba, São Paulo, CEP: 16015-050, Brazil.
| | - Vânia L R Matta
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| | - Márcia D Laurenti
- Laboratório De Patologia De Doenças Infecciosas, Faculdade De Medicina, Universidade De São Paulo, São Paulo, CEP: 01246903, Brazil.
| |
Collapse
|
4
|
Basu S, Nandy A, Biswas D. Keeping RNA polymerase II on the run: Functions of MLL fusion partners in transcriptional regulation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194563. [PMID: 32348849 DOI: 10.1016/j.bbagrm.2020.194563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/13/2020] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Since the identification of key MLL fusion partners as transcription elongation factors regulating expression of HOX cluster genes during hematopoiesis, extensive work from the last decade has resulted in significant progress in our overall mechanistic understanding of role of MLL fusion partner proteins in transcriptional regulation of diverse set of genes beyond just the HOX cluster. In this review, we are going to detail overall understanding of role of MLL fusion partner proteins in transcriptional regulation and thus provide mechanistic insights into possible MLL fusion protein-mediated transcriptional misregulation leading to aberrant hematopoiesis and leukemogenesis.
Collapse
Affiliation(s)
- Subham Basu
- Laboratory of Transcription Biology, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 32, India
| | - Arijit Nandy
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debabrata Biswas
- Laboratory of Transcription Biology, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 32, India.
| |
Collapse
|
5
|
Ichii O, Kimura J, Okamura T, Horino T, Nakamura T, Sasaki H, Elewa YHA, Kon Y. IL-36α Regulates Tubulointerstitial Inflammation in the Mouse Kidney. Front Immunol 2017; 8:1346. [PMID: 29109726 PMCID: PMC5660075 DOI: 10.3389/fimmu.2017.01346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/03/2017] [Indexed: 12/20/2022] Open
Abstract
IL-36α, a member of the IL-1 family, is a crucial mediator of inflammatory responses. We previously found that IL-36α was overexpressed in injured distal tubules (DTs); however, its pathological function remains unclear. Herein, unilateral ureter obstruction (UUO) or folic acid (FA) injection was performed in mouse kidneys to assess the role of IL-36α in kidney injury. IL-36α mRNA and protein expression significantly increased in the kidneys within 24 h after UUO. IL-36α localized to dilated DTs. IL-36α expression significantly correlated with the progression of tubulointerstitial cell infiltration and tubular epithelium cell death in UUO kidneys and with renal dysfunction in FA-induced acute kidney injury mice. At 24 h after UUO, IL-36α+ DT epithelial cells showed loose intercellular digitations. IL-1RL2, an IL-36α receptor protein, localized to podocytes, proximal tubules, and DTs in the healthy kidney. IL-1RL2 was expressed in interstitial cells and platelets or extended primary cilia of DT epithelial cells in UUO kidneys. IL-36α stimulation promoted the production of IL-6 and Prss35, an inflammatory cytokine and collagen remodeling-associated enzyme, respectively, in cultured NIH3T3 fibroblasts. UUO-treated IL-36α-knockout (KO) mice showed milder kidney injury features than wild-type (WT) mice did. In UUO kidneys from IL-36α-KO mice, the expression of genes associated with inflammatory response and sensory perception was significantly different from that in WT mice. Altogether, our data indicate an association between intrarenal IL-36α overexpression and the progression of tubulointerstitial inflammations and morpho-functional alterations of DT epithelial cells. IL-36α may be a novel kidney injury marker useful for evaluating DT damages.
Collapse
Affiliation(s)
- Osamu Ichii
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Department of Basic Veterinary Sciences, Hokkaido University, Sapporo, Japan
| | - Junpei Kimura
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Department of Basic Veterinary Sciences, Hokkaido University, Sapporo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, National Center for Global Health and Medicine, Tokyo, Japan.,Department of Infectious Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Taro Horino
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Teppei Nakamura
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Department of Basic Veterinary Sciences, Hokkaido University, Sapporo, Japan.,Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Chitose, Japan
| | - Hayato Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Yaser Hosny Ali Elewa
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Department of Basic Veterinary Sciences, Hokkaido University, Sapporo, Japan.,Faculty of Veterinary Medicine, Department of Histology and Cytology, Zagazig University, Zagazig, Egypt
| | - Yasuhiro Kon
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Department of Basic Veterinary Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
6
|
Abstract
Extracellular vesicles are a heterogeneous population of microparticles released by virtually all living cells which have been recently widely investigated in different biological fields. They are typically composed of two primary types (exosomes and microvesicles) and are recently commanding increasing attention as mediators of cellular signaling. Indeed, these vesicles can affect recipient cells by carrying and delivering complex cargos of biomolecules (including proteins, lipids and nucleic acids), protected from enzymatic degradation in the environment. Their importance has been demonstrated in the pathophysiology of several organs, in particular in kidney, where different cell types secrete extracellular vesicles that mediate their communication with downstream urinary tract cells. Over the past few years, evidence has been shown that vesicles participate in kidney development and normal physiology. Moreover, EVs are widely demonstrated to be implicated in cellular signaling during renal regenerative and pathological processes. Although many EV mechanisms are still poorly understood, in particular in kidney, the discovery of their role could help to shed light on renal biological processes which are so far elusive. Lastly, extracellular vesicles secreted by renal cells gather in urine, thus becoming a great resource for disease or recovery markers and a promising non-invasive diagnostic instrument for renal disease. In the present review, we discuss the most recent findings on the role of extracellular vesicles in renal physiopathology and their potential implication in diagnosis and therapy.
Collapse
Affiliation(s)
| | - Chiara Gai
- Stem Cell Laboratory, Department of Medical Sciences, University of TurinTurin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of TurinTurin, Italy
| | - Giovanni Camussi
- Stem Cell Laboratory, Department of Medical Sciences, University of TurinTurin, Italy
| |
Collapse
|
7
|
Mehra R, Vats P, Cieslik M, Cao X, Su F, Shukla S, Udager AM, Wang R, Pan J, Kasaian K, Lonigro R, Siddiqui J, Premkumar K, Palapattu G, Weizer A, Hafez KS, Wolf JS, Sangoi AR, Trpkov K, Osunkoya AO, Zhou M, Giannico G, McKenney JK, Dhanasekaran SM, Chinnaiyan AM. Biallelic Alteration and Dysregulation of the Hippo Pathway in Mucinous Tubular and Spindle Cell Carcinoma of the Kidney. Cancer Discov 2016; 6:1258-1266. [PMID: 27604489 DOI: 10.1158/2159-8290.cd-16-0267] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 09/02/2016] [Indexed: 11/16/2022]
Abstract
Mucinous tubular and spindle cell carcinoma (MTSCC) is a relatively rare subtype of renal cell carcinoma (RCC) with distinctive morphologic and cytogenetic features. Here, we carry out whole-exome and transcriptome sequencing of a multi-institutional cohort of MTSCC (n = 22). We demonstrate the presence of either biallelic loss of Hippo pathway tumor suppressor genes (TSG) and/or evidence of alteration of Hippo pathway genes in 85% of samples. PTPN14 (31%) and NF2 (22%) were the most commonly implicated Hippo pathway genes, whereas other genes such as SAV1 and HIPK2 were also involved in a mutually exclusive fashion. Mutations in the context of recurrent chromosomal losses amounted to biallelic alterations in these TSGs. As a readout of Hippo pathway inactivation, a majority of cases (90%) exhibited increased nuclear YAP1 protein expression. Taken together, nearly all cases of MTSCC exhibit some evidence of Hippo pathway dysregulation. SIGNIFICANCE MTSCC is a rare and relatively recently described subtype of RCC. Next-generation sequencing of a multi-institutional MTSCC cohort revealed recurrent chromosomal losses and somatic mutations in the Hippo signaling pathway genes leading to potential YAP1 activation. In virtually all cases of MTSCC, there was evidence of Hippo pathway dysregulation, suggesting a common mechanistic basis for this disease. Cancer Discov; 6(11); 1258-66. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1197.
Collapse
Affiliation(s)
- Rohit Mehra
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan Health System, Ann Arbor, Michigan.,Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Pankaj Vats
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan.,Michigan Center for Translational Pathology, Ann Arbor, Michigan.,Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Xuhong Cao
- Michigan Center for Translational Pathology, Ann Arbor, Michigan.,Howard Hughes Medical Institute, Ann Arbor, Michigan
| | - Fengyun Su
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Sudhanshu Shukla
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Aaron M Udager
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan
| | - Rui Wang
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Jincheng Pan
- Department of Urology, First Affiliated Hospital, Sun-Yat Sen University, Guangzhou, China
| | - Katayoon Kasaian
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Robert Lonigro
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Javed Siddiqui
- Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Kumpati Premkumar
- Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Ganesh Palapattu
- Department of Urology, University of Michigan Health System, Ann Arbor, Michigan
| | - Alon Weizer
- Comprehensive Cancer Center, University of Michigan Health System, Ann Arbor, Michigan.,Department of Urology, University of Michigan Health System, Ann Arbor, Michigan
| | - Khaled S Hafez
- Department of Urology, University of Michigan Health System, Ann Arbor, Michigan
| | - J Stuart Wolf
- Department of Urology, University of Michigan Health System, Ann Arbor, Michigan
| | - Ankur R Sangoi
- El Camino Hospital, Department of Pathology, Mountain View, California
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Adeboye O Osunkoya
- Departments of Pathology and Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Ming Zhou
- Department of Pathology, New York University School of Medicine, New York, New York
| | - Giovanna Giannico
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jesse K McKenney
- Cleveland Clinic, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland, Ohio
| | - Saravana M Dhanasekaran
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan.,Michigan Center for Translational Pathology, Ann Arbor, Michigan
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan. .,Comprehensive Cancer Center, University of Michigan Health System, Ann Arbor, Michigan.,Michigan Center for Translational Pathology, Ann Arbor, Michigan.,Howard Hughes Medical Institute, Ann Arbor, Michigan.,Department of Urology, University of Michigan Health System, Ann Arbor, Michigan
| |
Collapse
|
8
|
Chiabotto G, Bruno S, Collino F, Camussi G. Mesenchymal Stromal Cells Epithelial Transition Induced by Renal Tubular Cells-Derived Extracellular Vesicles. PLoS One 2016; 11:e0159163. [PMID: 27409796 PMCID: PMC4943710 DOI: 10.1371/journal.pone.0159163] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal-epithelial interactions play an important role in renal tubular morphogenesis and in maintaining the structure of the kidney. The aim of this study was to investigate whether extracellular vesicles (EVs) produced by human renal proximal tubular epithelial cells (RPTECs) may induce mesenchymal-epithelial transition of bone marrow-derived mesenchymal stromal cells (MSCs). To test this hypothesis, we characterized the phenotype and the RNA content of EVs and we evaluated the in vitro uptake and activity of EVs on MSCs. MicroRNA (miRNA) analysis suggested the possible implication of the miR-200 family carried by EVs in the epithelial commitment of MSCs. Bone marrow-derived MSCs were incubated with EVs, or RPTEC-derived total conditioned medium, or conditioned medium depleted of EVs. As a positive control, MSCs were co-cultured in a transwell system with RPTECs. Epithelial commitment of MSCs was assessed by real time PCR and by immunofluorescence analysis of cellular expression of specific mesenchymal and epithelial markers. After one week of incubation with EVs and total conditioned medium, we observed mesenchymal-epithelial transition in MSCs. Stimulation with conditioned medium depleted of EVs did not induce any change in mesenchymal and epithelial gene expression. Since EVs were found to contain the miR-200 family, we transfected MSCs using synthetic miR-200 mimics. After one week of transfection, mesenchymal-epithelial transition was induced in MSCs. In conclusion, miR-200 carrying EVs released from RPTECs induce the epithelial commitment of MSCs that may contribute to their regenerative potential. Based on experiments of MSC transfection with miR-200 mimics, we suggested that the miR-200 family may be involved in mesenchymal-epithelial transition of MSCs.
Collapse
Affiliation(s)
- Giulia Chiabotto
- Department of Medical Science, University of Torino, Medical School, Torino, Italy
| | - Stefania Bruno
- Department of Molecular Biotechnology and Healthy Science, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Collino
- Department of Medical Science, University of Torino, Medical School, Torino, Italy
| | - Giovanni Camussi
- Department of Medical Science, University of Torino, Medical School, Torino, Italy
- * E-mail:
| |
Collapse
|
9
|
Wang Y, Chaudhari S, Ren Y, Ma R. Impairment of hepatic nuclear factor-4α binding to the Stim1 promoter contributes to high glucose-induced upregulation of STIM1 expression in glomerular mesangial cells. Am J Physiol Renal Physiol 2015; 308:F1135-45. [PMID: 25786776 PMCID: PMC4437002 DOI: 10.1152/ajprenal.00563.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/16/2015] [Indexed: 11/22/2022] Open
Abstract
The present study was carried out to investigate if hepatic nuclear factor (HNF)4α contributed to the high glucose-induced increase in stromal interacting molecule (STIM)1 protein abundance in glomerular mesangial cells (MCs). Western blot and immunofluorescence experiments showed HNF4α expression in MCs. Knockdown of HNF4α using a small interfering RNA approach significantly increased mRNA expression levels of both STIM1 and Orai1 and protein expression levels of STIM1 in cultured human MCs. Consistently, overexpression of HNF4α reduced expressed STIM1 protein expression in human embryonic kidney-293 cells. Furthermore, high glucose treatment did not significantly change the abundance of HNF4α protein in MCs but significantly attenuated HNF4α binding activity to the Stim1 promoter. Moreover, knockdown of HNF4α significantly augmented store-operated Ca(2+) entry, which is known to be gated by STIM1 and has recently been found to be antifibrotic in MCs. In agreement with those results, knockdown of HNF4α significantly attenuated the fibrotic response of high glucose. These results suggest that HNF4α negatively regulates STIM1 transcription in MCs. High glucose increases STIM1 expression levels by impairing HNF4α binding activity to the Stim1 promoter, which subsequently releases Stim1 transcription from HNF4α repression. Since the STIM1-gated store-operated Ca(2+) entry pathway in MCs has an antifibrotic effect, inhibition of HNF4α in MCs might be a potential therapeutic option for diabetic kidney disease.
Collapse
Affiliation(s)
- Yanxia Wang
- Department of Integrative Physiology and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas; and
| | - Sarika Chaudhari
- Department of Integrative Physiology and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas; and
| | - Yuezhong Ren
- Department of Endocrinology, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, Zhejiang, China
| | - Rong Ma
- Department of Integrative Physiology and Cardiovascular Research Institute, University of North Texas Health Science Center, Fort Worth, Texas; and
| |
Collapse
|
10
|
Understanding the role of maternal diet on kidney development; an opportunity to improve cardiovascular and renal health for future generations. Nutrients 2015; 7:1881-905. [PMID: 25774605 PMCID: PMC4377888 DOI: 10.3390/nu7031881] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 01/08/2023] Open
Abstract
The leading causes of mortality and morbidity worldwide are cardiovascular disease (high blood pressure, high cholesterol and renal disease), cancer and diabetes. It is increasingly obvious that the development of these diseases encompasses complex interactions between adult lifestyle and genetic predisposition. Maternal malnutrition can influence the fetal and early life environment and pose a risk factor for the future development of adult diseases, most likely due to impaired organogenesis in the developing offspring. This then predisposes these offspring to cardiovascular disease and renal dysfunction in adulthood. Studies in experimental animals have further illustrated the significant impact maternal diet has on offspring health. Many studies report changes in kidney structure (a reduction in the number of nephrons in the kidney) in offspring of protein-deprived dams. Although the early studies suggested that increased blood pressure was also present in offspring of protein-restricted dams, this is not a universal finding and requires clarification. Importantly, to date, the literature offers little to no understanding of when in development these changes in kidney development occur, nor are the cellular and molecular mechanisms that drive these changes well characterised. Moreover, the mechanisms linking maternal nutrition and a suboptimal renal phenotype in offspring are yet to be discerned—one potential mechanism involves epigenetics. This review will focus on recent information on potential mechanisms by which maternal nutrition (focusing on malnutrition due to protein restriction, micronutrient restriction and excessive fat intake) influences kidney development and thereby function in later life.
Collapse
|
11
|
Menezes LF, Zhou F, Patterson AD, Piontek KB, Krausz KW, Gonzalez FJ, Germino GG. Network analysis of a Pkd1-mouse model of autosomal dominant polycystic kidney disease identifies HNF4α as a disease modifier. PLoS Genet 2012; 8:e1003053. [PMID: 23209428 PMCID: PMC3510057 DOI: 10.1371/journal.pgen.1003053] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 09/06/2012] [Indexed: 12/20/2022] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD; MIM ID's 173900, 601313, 613095) leads to end-stage kidney disease, caused by mutations in PKD1 or PKD2. Inactivation of Pkd1 before or after P13 in mice results in distinct early- or late-onset disease. Using a mouse model of ADPKD carrying floxed Pkd1 alleles and an inducible Cre recombinase, we intensively analyzed the relationship between renal maturation and cyst formation by applying transcriptomics and metabolomics to follow disease progression in a large number of animals induced before P10. Weighted gene co-expression network analysis suggests that Pkd1-cystogenesis does not cause developmental arrest and occurs in the context of gene networks similar to those that regulate/maintain normal kidney morphology/function. Knowledge-based Ingenuity Pathway Analysis (IPA) software identifies HNF4α as a likely network node. These results are further supported by a meta-analysis of 1,114 published gene expression arrays in Pkd1 wild-type tissues. These analyses also predict that metabolic pathways are key elements in postnatal kidney maturation and early steps of cyst formation. Consistent with these findings, urinary metabolomic studies show that Pkd1 cystic mutants have a distinct profile of excreted metabolites, with pathway analysis suggesting altered activity in several metabolic pathways. To evaluate their role in disease, metabolic networks were perturbed by inactivating Hnf4α and Pkd1. The Pkd1/Hnf4α double mutants have significantly more cystic kidneys, thus indicating that metabolic pathways could play a role in Pkd1-cystogenesis. Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common genetic cause of polycystic kidney disease and is responsible for 4.6% of the end-stage renal disease (ESRD) cases in the United States. It is most often caused by mutation in the PKD1 gene. To understand this disease, we made a mouse model in which we could delete the Pkd1 gene and study the animal as its kidney becomes cystic. Using this model, we had previously found that the maturation status of the animal determines whether cysts form within days or within months, and we had narrowed down this switch to a two-day interval. In the current study, we used the rapid cyst-forming model to analyze the expression pattern of thousands of genes in mutant and control kidneys, and metabolites excreted in the urine. Our results identify a number of genes that may be involved in cyst formation and suggest that metabolic changes may play a role in ADPKD and could alter disease progression. These analyses also predict that metabolic pathways are key elements in normal postnatal kidney maturation.
Collapse
Affiliation(s)
- Luis F. Menezes
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fang Zhou
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andrew D. Patterson
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Klaus B. Piontek
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kristopher W. Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gregory G. Germino
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
12
|
Wu B, Brooks JD. Gene expression changes induced by unilateral ureteral obstruction in mice. J Urol 2012; 188:1033-41. [PMID: 22819101 DOI: 10.1016/j.juro.2012.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Indexed: 11/29/2022]
Abstract
PURPOSE Loss of renal function is often the impetus for operative intervention in renal obstruction cases. Obstructive nephropathy is characterized by discrete morphological and physiological changes, including tubular dilatation, apoptosis and atrophy as well as interstitial cellular infiltration and progressive interstitial fibrosis. We hypothesized that gene expression alterations correlate with obstructive nephropathy and could serve as biomarkers for early intervention. MATERIALS AND METHODS C57BL/6 mice were subjected to unilateral ureteral obstruction or sham surgery at postnatal day 21. Kidneys were harvested 1, 2, 5 and 9 days postoperatively. RNA was extracted from kidneys and comprehensive gene expression profiling was performed with microarrays. IPA® pathway analysis software was used to analyze the biological function and gene networks of gene expression data. RESULTS Microarray analysis revealed more than 1,800 transcripts that were up-regulated or down-regulated during days 1 through 9 after obstruction, including many previously reported transcripts (FOS, CD44, CLU, SPP1 and EGF). Pathway analysis showed significant enrichment of transcripts in cell activation/differentiation, immune/inflammatory responses, cell cycle, metabolic process and transport. Network analysis using IPA showed that transcriptional regulatory pathways involving CEBPB and HNF4A are involved in obstructive nephropathy. CONCLUSIONS This data set provides a foundation for development of biomarkers for obstructive nephropathy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Urology, Stanford University School of Medicine, Stanford, California 94305-5118, USA
| | | |
Collapse
|
13
|
Gallegos TF, Martovetsky G, Kouznetsova V, Bush KT, Nigam SK. Organic anion and cation SLC22 "drug" transporter (Oat1, Oat3, and Oct1) regulation during development and maturation of the kidney proximal tubule. PLoS One 2012; 7:e40796. [PMID: 22808265 PMCID: PMC3396597 DOI: 10.1371/journal.pone.0040796] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 06/13/2012] [Indexed: 12/14/2022] Open
Abstract
Proper physiological function in the pre- and post-natal proximal tubule of the kidney depends upon the acquisition of selective permeability, apical-basolateral epithelial polarity and the expression of key transporters, including those involved in metabolite, toxin and drug handling. Particularly important are the SLC22 family of transporters, including the organic anion transporters Oat1 (originally identified as NKT) and Oat3 as well as the organic cation transporter Oct1. In ex vivo cultures of metanephric mesenchyme (MM; the embryonic progenitor tissue of the nephron) Oat function was evident before completion of nephron segmentation and corresponded with the maturation of tight junctions as measured biochemically by detergent extractability of the tight junction protein, ZO-1. Examination of available time series microarray data sets in the context of development and differentiation of the proximal tubule (derived from both in vivo and in vitro/ex vivo developing nephrons) allowed for correlation of gene expression data to biochemically and functionally defined states of development. This bioinformatic analysis yielded a network of genes with connectivity biased toward Hnf4α (but including Hnf1α, hyaluronic acid-CD44, and notch pathways). Intriguingly, the Oat1 and Oat3 genes were found to have strong temporal co-expression with Hnf4α in the cultured MM supporting the notion of some connection between the transporters and this transcription factor. Taken together with the ChIP-qPCR finding that Hnf4α occupies Oat1, Oat3, and Oct1 proximal promoters in the in vivo differentiating rat kidney, the data suggest a network of genes with Hnf4α at its center plays a role in regulating the terminal differentiation and capacity for drug and toxin handling by the nascent proximal tubule of the kidney.
Collapse
Affiliation(s)
- Thomas F. Gallegos
- Department of Pediatrics, University of California at San Diego, La Jolla, California, United States of America
| | - Gleb Martovetsky
- Department of Biomedical Sciences, University of California at San Diego, La Jolla, California, United States of America
| | - Valentina Kouznetsova
- Department of Medicine, University of California at San Diego, La Jolla, California, United States of America
| | - Kevin T. Bush
- Department of Pediatrics, University of California at San Diego, La Jolla, California, United States of America
| | - Sanjay K. Nigam
- Department of Pediatrics, University of California at San Diego, La Jolla, California, United States of America
- Department of Medicine, University of California at San Diego, La Jolla, California, United States of America
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
14
|
Pelletier L, Rebouissou S, Vignjevic D, Bioulac-Sage P, Zucman-Rossi J. HNF1α inhibition triggers epithelial-mesenchymal transition in human liver cancer cell lines. BMC Cancer 2011; 11:427. [PMID: 21975049 PMCID: PMC3203860 DOI: 10.1186/1471-2407-11-427] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 10/05/2011] [Indexed: 12/31/2022] Open
Abstract
Background Hepatocyte Nuclear Factor 1α (HNF1α) is an atypical homeodomain-containing transcription factor that transactivates liver-specific genes including albumin, α-1-antitrypsin and α- and β-fibrinogen. Biallelic inactivating mutations of HNF1A have been frequently identified in hepatocellular adenomas (HCA), rare benign liver tumors usually developed in women under oral contraceptives, and in rare cases of hepatocellular carcinomas developed in non-cirrhotic liver. HNF1α-mutated HCA (H-HCA) are characterized by a marked steatosis and show activation of glycolysis, lipogenesis, translational machinery and mTOR pathway. We studied the consequences of HNF1α silencing in hepatic cell lines, HepG2 and Hep3B and we reproduced most of the deregulations identified in H-HCA. Methods We transfected hepatoma cell lines HepG2 and Hep3B with siRNA targeting HNF1α and obtained a strong inhibition of HNF1α expression. We then looked at the phenotypic changes by microscopy and studied changes in gene expression using qRT-PCR and Western Blot. Results Hepatocytes transfected with HNF1α siRNA underwent severe phenotypic changes with loss of cell-cell contacts and development of migration structures. In HNF1α-inhibited cells, hepatocyte and epithelial markers were diminished and mesenchymal markers were over-expressed. This epithelial-mesenchymal transition (EMT) was related to the up regulation of several EMT transcription factors, in particular SNAIL and SLUG. We also found an overexpression of TGFβ1, an EMT initiator, in both cells transfected with HNF1α siRNA and H-HCA. Moreover, TGFβ1 expression is strongly correlated to HNF1α expression in cell models, suggesting regulation of TGFβ1 expression by HNF1α. Conclusion Our results suggest that HNF1α is not only important for hepatocyte differentiation, but has also a role in the maintenance of epithelial phenotype in hepatocytes.
Collapse
Affiliation(s)
- Laura Pelletier
- Inserm U674 Génomique fonctionnelle des tumeurs solides, Paris, France
| | | | | | | | | |
Collapse
|