1
|
Neves VCM, Satie Okajima L, Elbahtety E, Joseph S, Daly J, Menon A, Fan D, Volkyte A, Mainas G, Fung K, Dhami P, Pelegrine AA, Sharpe P, Nibali L, Ide M. Repurposing Metformin for periodontal disease management as a form of oral-systemic preventive medicine. J Transl Med 2023; 21:655. [PMID: 37814261 PMCID: PMC10563330 DOI: 10.1186/s12967-023-04456-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/19/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Despite the improvements in treatment over the last decades, periodontal disease (PD) affects millions of people around the world and the only treatment available is based on controlling microbial load. Diabetes is known to increase the risk of PD establishment and progression, and recently, glucose metabolism modulation by pharmaceutical or dietarian means has been emphasised as a significant modulator of non-communicable disease development. METHODS The impact of pharmaceutically controlling glucose metabolism in non-diabetic animals and humans (REBEC, UTN code: U1111-1276-1942) was investigated by repurposing Metformin, as a mean to manage periodontal disease and its associated systemic risk factors. RESULTS We found that glucose metabolism control via use of Metformin aimed at PD management resulted in significant prevention of bone loss during induced periodontal disease and age-related bone loss in vivo. Metformin also influenced the bacterial species present in the oral environment and impacted the metabolic epithelial and stromal responses to bacterial dysbiosis at a single cell level. Systemically, Metformin controlled blood glucose levels and age-related weight gain when used long-term. Translationally, our pilot randomized control trial indicated that systemic Metformin was safe to use in non-diabetic patients and affected the periodontal tissues. During the medication window, patients showed stable levels of systemic blood glucose, lower circulating hsCRP and lower insulin levels after periodontal treatment when compared to placebo. Finally, patients treated with Metformin had improved periodontal parameters when compared to placebo treated patients. CONCLUSION This is the first study to demonstrate that systemic interventions using Metformin in non-diabetic individuals aimed at PD prevention have oral-systemic effects constituting a possible novel form of preventive medicine for oral-systemic disease management.
Collapse
Affiliation(s)
- Vitor C M Neves
- Centre for Craniofacial and Regenerative Biology, FoDOCS, King's College London, London, UK.
- Periodontology Unit, Centre for Host-Microbiome Interactions, FoDOCS, King's College London, London, UK.
| | - Luciana Satie Okajima
- Department of Periodontology and Implantology, School of Dentistry, São Leopoldo Mandic, Campinas, Brazil
| | - Eyad Elbahtety
- Centre for Craniofacial and Regenerative Biology, FoDOCS, King's College London, London, UK
| | - Susan Joseph
- Periodontology Unit, Centre for Host-Microbiome Interactions, FoDOCS, King's College London, London, UK
| | - James Daly
- Centre for Craniofacial and Regenerative Biology, FoDOCS, King's College London, London, UK
| | - Athul Menon
- NIHR BRC Genomics Research Platform, Guy's and St Thomas' NHS Foundation Trust, King's College London School of Medicine, London, UK
| | - Di Fan
- Centre for Craniofacial and Regenerative Biology, FoDOCS, King's College London, London, UK
| | - Ayste Volkyte
- Periodontology Unit, Centre for Host-Microbiome Interactions, FoDOCS, King's College London, London, UK
| | - Giuseppe Mainas
- Periodontology Unit, Centre for Host-Microbiome Interactions, FoDOCS, King's College London, London, UK
| | - Kathy Fung
- NIHR BRC Genomics Research Platform, Guy's and St Thomas' NHS Foundation Trust, King's College London School of Medicine, London, UK
| | - Pawan Dhami
- NIHR BRC Genomics Research Platform, Guy's and St Thomas' NHS Foundation Trust, King's College London School of Medicine, London, UK
| | - Andre A Pelegrine
- Department of Periodontology and Implantology, School of Dentistry, São Leopoldo Mandic, Campinas, Brazil
| | - Paul Sharpe
- Centre for Craniofacial and Regenerative Biology, FoDOCS, King's College London, London, UK
- Institute of Animal Physiology and Genetics, Brno, Czech Republic
| | - Luigi Nibali
- Periodontology Unit, Centre for Host-Microbiome Interactions, FoDOCS, King's College London, London, UK
| | - Mark Ide
- Periodontology Unit, Centre for Host-Microbiome Interactions, FoDOCS, King's College London, London, UK
| |
Collapse
|
2
|
Landais Y, Vallot C. Multi-modal quantification of pathway activity with MAYA. Nat Commun 2023; 14:1668. [PMID: 36966153 PMCID: PMC10039856 DOI: 10.1038/s41467-023-37410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/16/2023] [Indexed: 03/27/2023] Open
Abstract
Signaling pathways can be activated through various cascades of genes depending on cell identity and biological context. Single-cell atlases now provide the opportunity to inspect such complexity in health and disease. Yet, existing reference tools for pathway scoring resume activity of each pathway to one unique common metric across cell types. Here, we present MAYA, a computational method that enables the automatic detection and scoring of the diverse modes of activation of biological pathways across cell populations. MAYA improves the granularity of pathway analysis by detecting subgroups of genes within reference pathways, each characteristic of a cell population and how it activates a pathway. Using multiple single-cell datasets, we demonstrate the biological relevance of identified modes of activation, the robustness of MAYA to noisy pathway lists and batch effect. MAYA can also predict cell types starting from lists of reference markers in a cluster-free manner. Finally, we show that MAYA reveals common modes of pathway activation in tumor cells across patients, opening the perspective to discover shared therapeutic vulnerabilities.
Collapse
Affiliation(s)
| | - Céline Vallot
- CNRS UMR3244, Institut Curie, PSL University, Paris, France.
- Translational Research Department, Institut Curie, PSL University, Paris, France.
- Single Cell Initiative, Institut Curie, PSL University, Paris, France.
| |
Collapse
|
3
|
Liu CC, Kim YJ, Teh R, Garcia A, Hamilton EJ, Cornelius F, Baxter RC, Rasmussen HH. Displacement of Native FXYD Protein From Na+/K+-ATPase With Novel FXYD Peptide Derivatives: Effects on Doxorubicin Cytotoxicity. Front Oncol 2022; 12:859216. [PMID: 35371992 PMCID: PMC8968713 DOI: 10.3389/fonc.2022.859216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
The seven mammalian FXYD proteins associate closely with α/β heterodimers of Na+/K+-ATPase. Most of them protect the β1 subunit against glutathionylation, an oxidative modification that destabilizes the heterodimer and inhibits Na+/K+-ATPase activity. A specific cysteine (Cys) residue of FXYD proteins is critical for such protection. One of the FXYD proteins, FXYD3, confers treatment resistance when overexpressed in cancer cells. We developed two FXYD3 peptide derivatives. FXYD3-pep CKCK retained the Cys residue that can undergo glutathionylation and that is critical for protecting the β1 subunit against glutathionylation. FXYD3-pep SKSK had all Cys residues mutated to Serine (Ser). The chemotherapeutic doxorubicin induces oxidative stress, and suppression of FXYD3 with siRNA in pancreatic- and breast cancer cells that strongly express FXYD3 increased doxorubicin-induced cytotoxicity. Exposing cells to FXYD3-pep SKSK decreased co-immunoprecipitation of FXYD3 with the α1 Na+/K+-ATPase subunit. FXYD3-pep SKSK reproduced the increase in doxorubicin-induced cytotoxicity seen after FXYD3 siRNA transfection in pancreatic- and breast cancer cells that overexpressed FXYD3, while FXYD3-pep CKCK boosted the native protein’s protection against doxorubicin. Neither peptide affected doxorubicin’s cytotoxicity on cells with no or low FXYD3 expression. Fluorescently labeled FXYD3-pep SKSK was detected in a perinuclear distribution in the cells overexpressing FXYD3, and plasmalemmal Na+/K+-ATPase turnover could not be implicated in the increased sensitivity to doxorubicin that FXYD3-pep SKSK caused. FXYD peptide derivatives allow rapid elimination or amplification of native FXYD protein function. Here, their effects implicate the Cys residue that is critical for countering β1 subunit glutathionylation in the augmentation of cytotoxicity with siRNA-induced downregulation of FXYD3.
Collapse
Affiliation(s)
- Chia-Chi Liu
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, St Leonards, NSW, Australia
- *Correspondence: Chia-Chi Liu, ; Helge H. Rasmussen,
| | - Yeon Jae Kim
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, St Leonards, NSW, Australia
| | - Rachel Teh
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, St Leonards, NSW, Australia
| | - Alvaro Garcia
- School of Chemistry, University of Sydney, Camperdown, NSW, Australia
| | - Elisha J. Hamilton
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, St Leonards, NSW, Australia
| | | | - Robert C. Baxter
- Hormones and Cancer Laboratories, Kolling Institute, University of Sydney, St Leonards, NSW, Australia
| | - Helge H. Rasmussen
- North Shore Heart Research Group, Kolling Medical Research Institute, University of Sydney, St Leonards, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, St Leonards, NSW, Australia
- *Correspondence: Chia-Chi Liu, ; Helge H. Rasmussen,
| |
Collapse
|
4
|
Limbad C, Doi R, McGirr J, Ciotlos S, Perez K, Clayton ZS, Daya R, Seals DR, Campisi J, Melov S. Senolysis induced by 25-hydroxycholesterol targets CRYAB in multiple cell types. iScience 2022; 25:103848. [PMID: 35198901 PMCID: PMC8851282 DOI: 10.1016/j.isci.2022.103848] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/24/2021] [Accepted: 01/25/2022] [Indexed: 01/18/2023] Open
Abstract
Cellular senescence is a driver of many age-related pathologies. There is an active search for pharmaceuticals termed senolytics that can mitigate or remove senescent cells in vivo by targeting genes that promote the survival of senescent cells. We utilized single-cell RNA sequencing to identify CRYAB as a robust senescence-induced gene and potential target for senolysis. Using chemical inhibitor screening for CRYAB disruption, we identified 25-hydroxycholesterol (25HC), an endogenous metabolite of cholesterol biosynthesis, as a potent senolytic. We then validated 25HC as a senolytic in mouse and human cells in culture and in vivo in mouse skeletal muscle. Thus, 25HC represents a potential class of senolytics, which may be useful in combating diseases or physiologies in which cellular senescence is a key driver.
Collapse
Affiliation(s)
| | - Ryosuke Doi
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Julia McGirr
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Kevin Perez
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Zachary S. Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Radha Daya
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Douglas R. Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA, USA
| |
Collapse
|
5
|
Bejček J, Spiwok V, Kmoníčková E, Rimpelová S. Na +/K +-ATPase Revisited: On Its Mechanism of Action, Role in Cancer, and Activity Modulation. Molecules 2021; 26:molecules26071905. [PMID: 33800655 PMCID: PMC8061769 DOI: 10.3390/molecules26071905] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
Maintenance of Na+ and K+ gradients across the cell plasma membrane is an essential process for mammalian cell survival. An enzyme responsible for this process, sodium-potassium ATPase (NKA), has been currently extensively studied as a potential anticancer target, especially in lung cancer and glioblastoma. To date, many NKA inhibitors, mainly of natural origin from the family of cardiac steroids (CSs), have been reported and extensively studied. Interestingly, upon CS binding to NKA at nontoxic doses, the role of NKA as a receptor is activated and intracellular signaling is triggered, upon which cancer cell death occurs, which lies in the expression of different NKA isoforms than in healthy cells. Two major CSs, digoxin and digitoxin, originally used for the treatment of cardiac arrhythmias, are also being tested for another indication—cancer. Such drug repositioning has a big advantage in smoother approval processes. Besides this, novel CS derivatives with improved performance are being developed and evaluated in combination therapy. This article deals with the NKA structure, mechanism of action, activity modulation, and its most important inhibitors, some of which could serve not only as a powerful tool to combat cancer, but also help to decipher the so-far poorly understood NKA regulation.
Collapse
Affiliation(s)
- Jiří Bejček
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic; (J.B.); (V.S.)
| | - Vojtěch Spiwok
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic; (J.B.); (V.S.)
| | - Eva Kmoníčková
- Department of Pharmacology, Second Faculty of Medicine, Charles University, Plzeňská 311, 150 00 Prague, Czech Republic;
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic; (J.B.); (V.S.)
- Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 323 00 Pilsen, Czech Republic
- Correspondence: ; Tel.: +420-220-444-360
| |
Collapse
|
6
|
Wang LJ, Li QJ, Le Y, Ouyang HY, He MK, Yu ZS, Zhang YF, Shi M. Prognostic significance of sodium-potassium ATPase regulator, FXYD3, in human hepatocellular carcinoma. Oncol Lett 2017; 15:3024-3030. [PMID: 29435033 PMCID: PMC5778849 DOI: 10.3892/ol.2017.7688] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 11/07/2017] [Indexed: 12/16/2022] Open
Abstract
The clinical significance of the sodium-potassium ATPase regulator FXYD domain-containing ion transport regulator 3 (FXYD3) has been demonstrated in a number of types of cancer. However, the role of this protein in human hepatocellular carcinoma (HCC) remains to be elucidated. In the present study, 217 HCC tissue samples were analyzed to evaluate the expression and prognostic significance of FXYD3 in HCC. Reverse transcription-quantitative polymerase chain reaction was used to analyze the mRNA expression of FXYD3 in 80 primary HCC specimens and paired non-cancerous liver tissue samples, while western blotting was used to analyze the protein expression level of FXYD3 in another 24 pairs. These analyses demonstrated that the expression level of FXYD3 was significantly increasedb at the mRNA and protein levels in HCC tumor tissues compared with adjacent non-cancerous tissues. Immunohistochemical analysis of 137 paraffin-embedded HCC tissue samples indicated that the expression of FXYD3 was associated with HCC clinicopathological characteristics. Kaplan-Meier analysis demonstrated that patients with high FXYD3 protein expression (n=60) experienced significantly poorer overall survival time compared with patients with low FXYD3 protein expression (n=77) (P<0.001). Multivariate analysis demonstrated that FYXD3 protein expression level (hazard ratio, 2.137; 95% confidence interval, 1.224–3.732; P=0.008) was an independent prognostic factor in patients with HCC. Overall, the results indicated that FXYD3 expression levels were higher in HCC tumor tissues than in adjacent non-cancerous tissues, and that the FXYD3 protein may serve as a prognostic marker for HCC.
Collapse
Affiliation(s)
- Li-Juan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Qi-Jiong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Yong Le
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Han-Yue Ouyang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Min-Ke He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zi-Shan Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Yong-Fa Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Ming Shi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
7
|
Wang PJ, Yang WK, Lin CH, Hwang HH, Lee TH. FXYD8, a Novel Regulator of Renal Na +/K +-ATPase in the Euryhaline Teleost, Tetraodon nigroviridis. Front Physiol 2017; 8:576. [PMID: 28848450 PMCID: PMC5550679 DOI: 10.3389/fphys.2017.00576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/25/2017] [Indexed: 11/19/2022] Open
Abstract
FXYD proteins are important regulators of Na+/K+-ATPase (NKA) activity in mammals. As an inhabitant of estuaries, the pufferfish (Tetraodon nigroviridis) responds to ambient salinity changes with efficient osmoregulation, including alterations in branchial, and renal NKA activities. Previous studies on teleostean FXYDs have mainly focused on the expression and potential functions of FXYD proteins in gills. The goal of the present study was to elucidate the potential role of FXYD8, a member of the fish FXYD protein family, in the modulation of NKA activity in the kidneys of this euryhaline pufferfish by using molecular, biochemical, and physiological approaches. The results demonstrate that T. nigroviridis FXYD8 (TnFXYD8) interacts with NKA in renal tubules. Meanwhile, the protein expression of renal TnFXYD8 was found to be significantly upregulated in hyperosmotic seawater-acclimated pufferfish. Moreover, overexpression of TnFXYD8 in Xenopus oocytes decreased NKA activity. Our results suggest the FXYD8 is able to modulate NKA activity through inhibitory effects upon salinity challenge. The present study further extends our understanding of the functions of FXYD proteins, the regulators of NKA, in vertebrates.
Collapse
Affiliation(s)
- Pei-Jen Wang
- Department of Life Sciences, National Chung Hsing UniversityTaichung, Taiwan
- Department of Public Affairs and Civic Education, National Changhua University of EducationChanghua, Taiwan
| | - Wen-Kai Yang
- Department of Life Sciences, National Chung Hsing UniversityTaichung, Taiwan
- Bachelor Degree Program in Animal Healthcare, Hungkuang UniversityTaichung, Taiwan
| | - Chia-Hao Lin
- National Institute for Basic Biology, National Institutes of Natural SciencesOkazaki, Japan
| | - Hau-Hsuan Hwang
- Department of Life Sciences, National Chung Hsing UniversityTaichung, Taiwan
| | - Tsung-Han Lee
- Department of Life Sciences, National Chung Hsing UniversityTaichung, Taiwan
- Agricultural Biotechnology Center, National Chung Hsing UniversityTaichung, Taiwan
| |
Collapse
|
8
|
Hou F, Huang QM, Hu DN, Jonas JB, Wei WB. Immune oppression array elucidating immune escape and survival mechanisms in uveal melanoma. Int J Ophthalmol 2016; 9:1701-1712. [PMID: 28003967 DOI: 10.18240/ijo.2016.12.01] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/27/2016] [Indexed: 12/18/2022] Open
Abstract
AIM To examine the genetic profile of primary uveal melanoma (UM) as compared to UM in immune escape. METHODS Dendritic cells (DC) loaded with lysates of UM cells of high metastatic potential were used to stimulate CTLs(CTLs). When CTLs co-cultured with the UM cells, most UM cells could be eliminated. Survival UM cells grew slowly and were considered to be survival variants and examined by a microarray analysis. These differential genes were analyzed further with Venn Diagrams and functions related to immune escape. We additionally examined transcriptional changes of manually selected survival variants of UM cells and of clinical UM samples by quantitative real-time polymerase chain reaction (qRT-PCR), and analyzed the correlation of these expressions and patients' survival. RESULTS Gene expression analyses revealed a marked up-regulation of SLAMF7 and CCL22 and a significant down-regulation of KRT10, FXYD3 and ABCC2. The expression of these genes in the relapsed UM was significantly greater than those in primary UM. UM patients with overexpression of these genes had a shorter survival period as compared with those of their underexpression. CONCLUSION Gene expression, in particular of SLAMF7, CCL22, KRT10, FXYD3 and ABCC2, differed between primary UM cells and survival variants of UM cells.
Collapse
Affiliation(s)
- Fang Hou
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Lab, Beijing 100730, China
| | - Qi-Ming Huang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Lab, Beijing 100730, China
| | - Dan-Ning Hu
- Departments of Ophthalmology and Pathology, New York Eye and Ear Infirmary of Mount Sinai, 310 E.14th St., NY 10003, USA
| | - Jost B Jonas
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Lab, Beijing 100730, China; Department of Ophthalmology, Medical Faculty Mannheim of the Ruprecht-Karls-University, Heidelberg 67117, Germany
| | - Wen-Bin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Lab, Beijing 100730, China
| |
Collapse
|
9
|
Wujak ŁA, Blume A, Baloğlu E, Wygrecka M, Wygowski J, Herold S, Mayer K, Vadász I, Besuch P, Mairbäurl H, Seeger W, Morty RE. FXYD1 negatively regulates Na(+)/K(+)-ATPase activity in lung alveolar epithelial cells. Respir Physiol Neurobiol 2015; 220:54-61. [PMID: 26410457 DOI: 10.1016/j.resp.2015.09.008] [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: 05/08/2015] [Revised: 09/17/2015] [Accepted: 09/20/2015] [Indexed: 01/10/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is clinical syndrome characterized by decreased lung fluid reabsorption, causing alveolar edema. Defective alveolar ion transport undertaken in part by the Na(+)/K(+)-ATPase underlies this compromised fluid balance, although the molecular mechanisms at play are not understood. We describe here increased expression of FXYD1, FXYD3 and FXYD5, three regulatory subunits of the Na(+)/K(+)-ATPase, in the lungs of ARDS patients. Transforming growth factor (TGF)-β, a pathogenic mediator of ARDS, drove increased FXYD1 expression in A549 human lung alveolar epithelial cells, suggesting that pathogenic TGF-β signaling altered Na(+)/K(+)-ATPase activity in affected lungs. Lentivirus-mediated delivery of FXYD1 and FXYD3 allowed for overexpression of both regulatory subunits in polarized H441 cell monolayers on an air/liquid interface. FXYD1 but not FXYD3 overexpression inhibited amphotericin B-sensitive equivalent short-circuit current in Ussing chamber studies. Thus, we speculate that FXYD1 overexpression in ARDS patient lungs may limit Na(+)/K(+)-ATPase activity, and contribute to edema persistence.
Collapse
Affiliation(s)
- Łukasz A Wujak
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Biochemistry, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Anna Blume
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Emel Baloğlu
- Department of Sports Medicine, Medical Clinic VII, University Hospital Heidelberg, University of Heidelberg, Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany; Department of Medical Pharmacology, Acibadem University, İstanbul, Turkey
| | - Małgorzata Wygrecka
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Biochemistry, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jegor Wygowski
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Konstantin Mayer
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Petra Besuch
- Department of Pathology, Klinikum Frankfurt (Oder) GmbH, Frankfurt (Oder), Germany
| | - Heimo Mairbäurl
- Department of Biochemistry, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rory E Morty
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| |
Collapse
|
10
|
Vallois D, Niederhäuser G, Ibberson M, Nagaray V, Marselli L, Marchetti P, Chatton JY, Thorens B. Gluco-incretins regulate beta-cell glucose competence by epigenetic silencing of Fxyd3 expression. PLoS One 2014; 9:e103277. [PMID: 25058609 PMCID: PMC4110016 DOI: 10.1371/journal.pone.0103277] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 06/28/2014] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND/AIMS Gluco-incretin hormones increase the glucose competence of pancreatic beta-cells by incompletely characterized mechanisms. METHODS We searched for genes that were differentially expressed in islets from control and Glp1r-/-; Gipr-/- (dKO) mice, which show reduced glucose competence. Overexpression and knockdown studies; insulin secretion analysis; analysis of gene expression in islets from control and diabetic mice and humans as well as gene methylation and transcriptional analysis were performed. RESULTS Fxyd3 was the most up-regulated gene in glucose incompetent islets from dKO mice. When overexpressed in beta-cells Fxyd3 reduced glucose-induced insulin secretion by acting downstream of plasma membrane depolarization and Ca++ influx. Fxyd3 expression was not acutely regulated by cAMP raising agents in either control or dKO adult islets. Instead, expression of Fxyd3 was controlled by methylation of CpGs present in its proximal promoter region. Increased promoter methylation reduced Fxyd3 transcription as assessed by lower abundance of H3K4me3 at the transcriptional start site and in transcription reporter assays. This epigenetic imprinting was initiated perinatally and fully established in adult islets. Glucose incompetent islets from diabetic mice and humans showed increased expression of Fxyd3 and reduced promoter methylation. CONCLUSIONS/INTERPRETATION Because gluco-incretin secretion depends on feeding the epigenetic regulation of Fxyd3 expression may link nutrition in early life to establishment of adult beta-cell glucose competence; this epigenetic control is, however, lost in diabetes possibly as a result of gluco-incretin resistance and/or de-differentiation of beta-cells that are associated with the development of type 2 diabetes.
Collapse
Affiliation(s)
- David Vallois
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Guy Niederhäuser
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Mark Ibberson
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- Vital-IT group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Lorella Marselli
- Department of Endocrinology and Metabolism, Ospedale di Cisanello, Pisa, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, Ospedale di Cisanello, Pisa, Italy
| | - Jean-Yves Chatton
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
11
|
Overexpression of FXYD-3 is involved in the tumorigenesis and development of esophageal squamous cell carcinoma. DISEASE MARKERS 2013; 35:195-202. [PMID: 24167366 PMCID: PMC3774971 DOI: 10.1155/2013/740201] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 06/13/2013] [Accepted: 07/16/2013] [Indexed: 01/03/2023]
Abstract
Objective. To investigate the association of FXYD-3 expression with clinicopathological variables and PINCH in patients with ESCC. Patients and Methods. Expression of FXYD-3 protein was immunohistochemically examined in normal esophageal mucous (n = 20) and ESCC (n = 64). Results. Expression of FXYD-3 in the cytoplasm markedly increased from normal esophageal epithelial cells to primary ESCC (P = 0.001). The expression of FXYD-3 was correlated with TNM stages and depth of tumor invasion. Furthermore, the cases with lymph node metastasis tended to show a higher frequency of positive expression than those without metastasis (P = 0.086), and FXYD-3 expression tended to be positively related to the expression of PINCH (P = 0.063). Moreover, the cases positive for both proteins had the highest frequency of lymph node metastasis (P = 0.001). However, FXYD-3 expression was not correlated with patient's gender (P = 0.847), age (P = 0.876), tumor location (P = 0.279), size (P = 0.771), grade of differentiation (P = 0.279), and survival (P = 0.113). Conclusion. Overexpression of FXYD-3 in the cytoplasm may play an important role in the tumorigenesis and development in the human ESCC, particularly in combination with PINCH expression.
Collapse
|
12
|
Yamamoto H, Mukaisho KI, Sugihara H, Hattori T, Asano S. Down-regulation of FXYD3 is induced by transforming growth factor-β signaling via ZEB1/δEF1 in human mammary epithelial cells. Biol Pharm Bull 2011; 34:324-9. [PMID: 21372379 DOI: 10.1248/bpb.34.324] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
FXYD3, a regulator of Na, K-ATPase, was identified as an mRNA overexpressed in murine breast cancers induced by neu oncogene, which had inactivated transforming growth factor (TGF)-β signaling due to the defect of TGF-β receptor I (TβRI) expression. To elucidate whether the expression of FXYD3 mRNA was regulated by TGF-β signaling, we used a normal human mammary epithelial cell line, MCF-10A which responds to TGF-β and tumor necrosis factor (TNF)-α, followed by induction of epithelial-to-mesenchymal transition (EMT). Here, we showed that FXYD3 at plasma membrane in epithelial state of MCF-10A cells was decreased by treatment of TGF-β and TNF-α. The repression of FXYD3 mRNA induced by TGF-β and TNF-α in MCF-10A cells was abolished by TβRI inhibitor or Smad3 inhibitor, but not by small interfering RNA (siRNA) for Smad2. In addition, expression level of FXYD3 mRNA was up-regulated by the silencing of ZEB1/δEF1 transcriptional repressor which was a down-stream target gene of TGF-β and an inducer of EMT. On the other hand, expression level and cellular localization of E-cadherin and N-cadherin were not changed by siRNA for FXYD3 in MCF-10A and human breast cancer MCF-7 cells. These results suggest that FXYD3 is a target gene of TGF-β signaling through ZEB1/δEF1, but is not directly involved in EMT.
Collapse
Affiliation(s)
- Hiroto Yamamoto
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga 525–8577, Japan
| | | | | | | | | |
Collapse
|
13
|
Lubarski I, Asher C, Garty H. FXYD5 (dysadherin) regulates the paracellular permeability in cultured kidney collecting duct cells. Am J Physiol Renal Physiol 2011; 301:F1270-80. [PMID: 21900457 DOI: 10.1152/ajprenal.00142.2011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
FXYD5 (dysadherin or RIC) is a member of the FXYD family of single-span transmembrane proteins associated with the Na(+)-K(+)-ATPase. Several studies have demonstrated enhanced expression of FXYD5 during metastasis and effects on cell adhesion and motility. The current study examines effects of FXYD5 on the paracellular permeability in the mouse kidney collecting duct cell line M1. Expressing FXYD5 in these cells leads to a large decrease in amiloride-insensitive transepithelial electrical resistance as well as increased permeability to 4-kDa dextran. Impairment of cell-cell contact was also demonstrated by staining cells for the tight and adherence junction markers zonula occludens-1 and β-catenin, respectively. This is further supported by large expansions of the interstitial spaces, visualized in electron microscope images. Expressing FXYD5 in M1 cells resulted in a decrease in N-glycosylation of β1 Na(+)-K(+)-ATPase, while silencing it in H1299 cells had an opposite effect. This may provide a mechanism for the above effects, since normal glycosylation of β1 plays an important role in cell-cell contact formation (Vagin O, Tokhtaeva E, Sachs G. J Biol Chem 281: 39573-39587, 2006).
Collapse
Affiliation(s)
- Irina Lubarski
- Dept. of Biological Chemistry, The Weizmann Institute of Science, Israel
| | | | | |
Collapse
|
14
|
Pusch J, Votteler M, Göhler S, Engl J, Hampel M, Walles H, Schenke-Layland K. The physiological performance of a three-dimensional model that mimics the microenvironment of the small intestine. Biomaterials 2011; 32:7469-78. [PMID: 21764120 DOI: 10.1016/j.biomaterials.2011.06.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 06/14/2011] [Indexed: 01/10/2023]
Abstract
Our focus was to develop a three-dimensional (3D) human dynamic in vitro tissue model that mimics the natural microenvironment of the small intestine. We co-cultured human Caco-2 cells with primary-isolated human microvascular endothelial cells (hMECs) on decellularized porcine jejunal segments within a custom-made dynamic bioreactor system that resembles the apical and basolateral side of the intestine for up to 14 days. The obtained data were compared to results generated using routine static Caco-2 assays. We performed histology and immunohistochemistry. Permeability was measured using directed transport studies. Histological analyses revealed that in tissue-engineered segments, which had been cultured under dynamic conditions, the Caco-2 cells showed a high-prismatic morphology, resembling normal primary enterocytes within their native environment. We further identified that the transport of low permeable substances, such as fluorescein and desmopressin increased within the dynamic bioreactor cultures. Immunohistochemical staining showed a significantly higher expression of the efflux transport p-glycoprotein (p-gp) under dynamic culture conditions when compared to the static cultures. We conclude that the integration of physiological parameters is crucial for the establishment of a reliable 3D intestinal in vitro model, which enables the simulation of drug transport over the gut-blood-barrier in a simplified way.
Collapse
Affiliation(s)
- Jacqueline Pusch
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Department of Cell and Tissue Engineering, Stuttgart, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Bachmann O, Juric M, Seidler U, Manns MP, Yu H. Basolateral ion transporters involved in colonic epithelial electrolyte absorption, anion secretion and cellular homeostasis. Acta Physiol (Oxf) 2011; 201:33-46. [PMID: 20528802 DOI: 10.1111/j.1748-1716.2010.02153.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Electrolyte transporters located in the basolateral membrane of the colonic epithelium are increasingly appreciated as elaborately regulated components of specific transport functions and cellular homeostasis: During electrolyte absorption, Na(+) /K(+) ATPase, Cl⁻ conductance, Cl⁻/HCO₃⁻ exchange, K(+) /Cl⁻ cotransport and K(+) channels are candidates for basolateral Na(+) , Cl⁻ and K(+) extrusion. The process of colonic anion secretion involves basolateral Na(+) /K(+) /2Cl⁻ , and probably also Na(+) /HCO₃⁻ cotransport, as well as Na(+) /K(+) ATPase and K(+) channels to supply substrate, stabilize the membrane potential and generate driving force respectively. Together with a multitude of additional transport systems, Na(+) /H(+) exchange and Na(+) /HCO₃⁻ cotransport have been implicated in colonocyte pH(i) and volume homeostasis. The purpose of this article is to summarize recently gathered information on the molecular identity, function and regulation of the involved basolateral transport systems in native tissue. Furthermore, we discuss how these findings can help to integrate these systems into the transport function and the cellular homoeostasis of colonic epithelial cells. Finally, disturbances of basolateral electrolyte transport during disease states such as mucosal inflammation will be reviewed.
Collapse
Affiliation(s)
- O Bachmann
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Germany.
| | | | | | | | | |
Collapse
|
16
|
Iwata Y, Yamada K, Iwayama Y, Anitha A, Thanseem I, Toyota T, Hattori E, Ohnishi T, Maekawa M, Nakamura K, Suzuki K, Matsuzaki H, Tsuchiya KJ, Suda S, Sugihara G, Takebayashi K, Yamamoto S, Iwata K, Mori N, Yoshikawa T. Failure to confirm genetic association of the FXYD6 gene with schizophrenia: the Japanese population and meta-analysis. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:1221-7. [PMID: 20468077 DOI: 10.1002/ajmg.b.31095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The FXYD domain-containing ion transport regulator 6 (FXYD6) gene encodes phosphohippolin that regulates cellular ion transport by altering the kinetic properties of Na,K-ATPase. Phosphohippolin is highly expressed in brain regions that are relevant to schizophrenia. The FXYD6 gene is located at chromosome 11q22-24, one of the most established linkage regions for schizophrenia. Therefore, it may be possible that genetic variants in FXYD6, including the regulatory genomic elements could cause abnormal function or expression of phosphohippolin and increase the genetic risk for schizophrenia. A previous study suggested that polymorphisms in FXYD6 are associated with schizophrenia in UK samples. However, conflicting results have been reported in the Japanese population. In this study, we aimed to test the prior genetic association findings using different samples from the ethnically homogeneous Japanese population (1,060 schizophrenic patients and 1,060 age- and sex-matched controls). From the FXYD6 gene, we examined six single nucleotide polymorphisms (rs11216573, rs555577, rs1815774, rs4938445, rs4938446, and rs497768), all of which were previously analyzed for association. We did not detect any significant allelic, genotypic or haplotypic association in our Japanese samples. Meta-analysis incorporating previous and the present studies also showed that the FXYD6 gene is not associated with schizophrenia. We conclude that the FXYD6 gene does not have a major influence on susceptibility to schizophrenia across populations.
Collapse
Affiliation(s)
- Yasuhide Iwata
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Translocation of Pseudomonas aeruginosa from the intestinal tract is mediated by the binding of ExoS to an Na,K-ATPase regulator, FXYD3. Infect Immun 2010; 78:4511-22. [PMID: 20805335 DOI: 10.1128/iai.00428-10] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The intestinal tract is considered the most important reservoir of Pseudomonas aeruginosa in intensive care units (ICUs). Gut colonization by P. aeruginosa underlies the development of invasive infections such as gut-derived sepsis. Intestinal colonization by P. aeruginosa is associated with higher ICU mortality rates. The translocation of endogenous P. aeruginosa from the colonized intestinal tract is an important pathogenic phenomenon. Here we identify bacterial and host proteins associated with bacterial penetration through the intestinal epithelial barrier. We first show by comparative genomic hybridization analysis that the exoS gene, encoding the type III effector protein, ExoS, was specifically detected in a clinical isolate that showed higher virulence in silkworms following midgut injection. We further show using a silkworm oral infection model that exoS is required both for virulence and for bacterial translocation from the midgut to the hemolymph. Using a bacterial two-hybrid screen, we show that the mammalian factor FXYD3, which colocalizes with and regulates the function of Na,K-ATPase, directly binds ExoS. A pulldown assay revealed that ExoS binds to the transmembrane domain of FXYD3, which also interacts with Na,K-ATPase. Na,K-ATPase controls the structure and barrier function of tight junctions in epithelial cells. Collectively, our results suggest that ExoS facilitates P. aeruginosa penetration through the intestinal epithelial barrier by binding to FXYD3 and thereby impairing the defense function of tight junctions against bacterial penetration.
Collapse
|