Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 43(1): 27, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38254102

ABSTRACT

BACKGROUND: Peritoneal metastasis, which accounts for 85% of all epithelial ovarian carcinoma (EOC) metastases, is a multistep process that requires the establishment of adhesive interactions between cancer cells and the peritoneal membrane. Interrelations between EOC and the mesothelial stroma are critical to facilitate the metastatic process. No data is available so far on the impact of histone acetylation/deacetylation, a potentially relevant mechanism governing EOC metastasis, on mesothelial cells (MCs)-mediated adhesion. METHODS: Static adhesion and peritoneal clearance experiments were performed pretreating mesenchymal-like MCs and platinum-sensitive/resistant EOC cell lines with MS-275-a Histone deacetylase (HDAC)1-3 pharmacological inhibitor currently used in combination trials. Results were acquired by confocal microscopy and were analyzed with an automated Opera software. The role of HDAC1/2 was validated by genetic silencing. The role of α4-, α5-α1 Integrins and Fibronectin-1 was validated using specific monoclonal antibodies. Quantitative proteomic analysis was performed on primary MCs pretreated with MS-275. Decellularized matrices were generated from either MS-275-exposed or untreated cells to study Fibronectin-1 extracellular secretion. The effect of MS-275 on ß1 integrin activity was assessed using specific monoclonal antibodies. The role of Talin-1 in MCs/EOC adhesion was analyzed by genetic silencing. Talin-1 ectopic expression was validated as a rescue tool from MS-275-induced phenotype. The in vivo effect of MS-275-induced MC remodeling was validated in a mouse model of peritoneal EOC dissemination. RESULTS: Treatment of MCs with non-cytotoxic concentrations of MS-275 caused a consistent reduction of EOC adhesion. Proteomic analysis revealed several pathways altered upon MC treatment with MS-275, including ECM deposition/remodeling, adhesion receptors and actin cytoskeleton regulators. HDAC1/2 inhibition hampered actin cytoskeleton polymerization by downregulating actin regulators including Talin-1, impairing ß1 integrin activation, and leading to abnormal extracellular secretion and distribution of Fibronectin-1. Talin-1 ectopic expression rescued EOC adhesion to MS-275-treated MCs. In an experimental mouse model of metastatic EOC, MS-275 limited tumor invasion, Fibronectin-1 secretion and the sub-mesothelial accumulation of MC-derived carcinoma-associated fibroblasts. CONCLUSION: Our study unveils a direct impact of HDAC-1/2 in the regulation of MC/EOC adhesion and highlights the regulation of MC plasticity by epigenetic inhibition as a potential target for therapeutic intervention in EOC peritoneal metastasis.


Subject(s)
Benzamides , Carcinoma, Ovarian Epithelial , Cell Adhesion , Histone Deacetylase 1 , Histone Deacetylase 2 , Ovarian Neoplasms , Peritoneal Neoplasms , Animals , Female , Humans , Mice , Actin Cytoskeleton/metabolism , Antibodies, Monoclonal , Carcinoma, Ovarian Epithelial/metabolism , Epithelium , Extracellular Matrix Proteins/metabolism , Fibronectins , Histone Deacetylase 1/metabolism , Integrin alpha5 , Integrin beta1/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Proteomics , Pyridines , Talin/genetics , Talin/metabolism , Histone Deacetylase 2/metabolism , Cell Adhesion/genetics
2.
Front Cell Dev Biol ; 10: 901321, 2022.
Article in English | MEDLINE | ID: mdl-35756995

ABSTRACT

Lipid droplets (LDs) are spherical, single sheet phospholipid-bound organelles that store neutral lipids in all eukaryotes and some prokaryotes. Initially conceived as relatively inert depots for energy and lipid precursors, these highly dynamic structures play active roles in homeostatic functions beyond metabolism, such as proteostasis and protein turnover, innate immunity and defense. A major share of the knowledge behind this paradigm shift has been enabled by the use of systematic molecular profiling approaches, capable of revealing and describing these non-intuitive systems-level relationships. Here, we discuss these advances and some of the challenges they entail, and highlight standing questions in the field.

3.
Cells ; 10(8)2021 08 05.
Article in English | MEDLINE | ID: mdl-34440759

ABSTRACT

Cells and tissues are continuously exposed to both chemical and physical stimuli and dynamically adapt and respond to this variety of external cues to ensure cellular homeostasis, regulated development and tissue-specific differentiation. Alterations of these pathways promote disease progression-a prominent example being cancer. Rho GTPases are key regulators of the remodeling of cytoskeleton and cell membranes and their coordination and integration with different biological processes, including cell polarization and motility, as well as other signaling networks such as growth signaling and proliferation. Apart from the control of GTP-GDP cycling, Rho GTPase activity is spatially and temporally regulated by post-translation modifications (PTMs) and their assembly onto specific protein complexes, which determine their controlled activity at distinct cellular compartments. Although Rho GTPases were traditionally conceived as targeted from the cytosol to the plasma membrane to exert their activity, recent research demonstrates that active pools of different Rho GTPases also localize to endomembranes and the nucleus. In this review, we discuss how PTM-driven modulation of Rho GTPases provides a versatile mechanism for their compartmentalization and functional regulation. Understanding how the subcellular sorting of active small GTPase pools occurs and what its functional significance is could reveal novel therapeutic opportunities.


Subject(s)
Protein Processing, Post-Translational , rho GTP-Binding Proteins/metabolism , Animals , Humans , Isoenzymes , Protein Transport , Signal Transduction
4.
Front Immunol ; 12: 671485, 2021.
Article in English | MEDLINE | ID: mdl-33981316

ABSTRACT

ECM composition and architecture are tightly regulated for tissue homeostasis. Different disorders have been associated to alterations in the levels of proteins such as collagens, fibronectin (FN) or tenascin-C (TnC). TnC emerges as a key regulator of multiple inflammatory processes, both during physiological tissue repair as well as pathological conditions ranging from tumor progression to cardiovascular disease. Importantly, our current understanding as to how TnC and other non-collagen ECM components are secreted has remained elusive. Extracellular vesicles (EVs) are small membrane-bound particles released to the extracellular space by most cell types, playing a key role in cell-cell communication. A broad range of cellular components can be transported by EVs (e.g. nucleic acids, lipids, signalling molecules and proteins). These cargoes can be transferred to target cells, potentially modulating their function. Recently, several extracellular matrix (ECM) proteins have been characterized as bona fide EV cargoes, exosomal secretion being particularly critical for TnC. EV-dependent ECM secretion might underpin diseases where ECM integrity is altered, establishing novel concepts in the field such as ECM nucleation over long distances, and highlighting novel opportunities for diagnostics and therapeutic intervention. Here, we review recent findings and standing questions on the molecular mechanisms governing EV-dependent ECM secretion and its potential relevance for disease, with a focus on TnC.


Subject(s)
Extracellular Matrix/metabolism , Extracellular Vesicles/metabolism , Tenascin/metabolism , Animals , Humans
5.
J Cell Biol ; 219(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-33053168

ABSTRACT

The composition and physical properties of the extracellular matrix (ECM) critically influence tumor progression, but the molecular mechanisms underlying ECM layering are poorly understood. Tumor-stroma interaction critically depends on cell communication mediated by exosomes, small vesicles generated within multivesicular bodies (MVBs). We show that caveolin-1 (Cav1) centrally regulates exosome biogenesis and exosomal protein cargo sorting through the control of cholesterol content at the endosomal compartment/MVBs. Quantitative proteomics profiling revealed that Cav1 is required for exosomal sorting of ECM protein cargo subsets, including Tenascin-C (TnC), and for fibroblast-derived exosomes to efficiently deposit ECM and promote tumor invasion. Cav1-driven exosomal ECM deposition not only promotes local stromal remodeling but also the generation of distant ECM-enriched stromal niches in vivo. Cav1 acts as a cholesterol rheostat in MVBs, determining sorting of ECM components into specific exosome pools and thus ECM deposition. This supports a model by which Cav1 is a central regulatory hub for tumor-stroma interactions through a novel exosome-dependent ECM deposition mechanism.


Subject(s)
Caveolin 1/physiology , Exosomes/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Multivesicular Bodies/metabolism , Proteome/metabolism , Tenascin/physiology , Animals , Fibroblasts/cytology , Mice , Mice, Knockout
6.
Cell Death Dis ; 11(8): 647, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32811813

ABSTRACT

Despite their emerging relevance to fully understand disease pathogenesis, we have as yet a poor understanding as to how biomechanical signals are integrated with specific biochemical pathways to determine cell behaviour. Mesothelial-to-mesenchymal transition (MMT) markers colocalized with TGF-ß1-dependent signaling and yes-associated protein (YAP) activation across biopsies from different pathologies exhibiting peritoneal fibrosis, supporting mechanotransduction as a central driving component of these class of fibrotic lesions and its crosstalk with specific signaling pathways. Transcriptome and proteome profiling of the response of mesothelial cells (MCs) to linear cyclic stretch revealed molecular changes compatible with bona fide MMT, which (i) overlapped with established YAP target gene subsets, and were largely dependent on endogenous TGF-ß1 signaling. Importantly, TGF-ß1 blockade blunts the transcriptional upregulation of these gene signatures, but not the mechanical activation and nuclear translocation of YAP per se. We studied the role therein of caveolin-1 (CAV1), a plasma membrane mechanotransducer. Exposure of CAV1-deficient MCs to cyclic stretch led to a robust upregulation of MMT-related gene programs, which was blunted upon TGF-ß1 inhibition. Conversely, CAV1 depletion enhanced both TGF-ß1 and TGFBRI expression, whereas its re-expression blunted mechanical stretching-induced MMT. CAV1 genetic deficiency exacerbated MMT and adhesion formation in an experimental murine model of peritoneal ischaemic buttons. Taken together, these results support that CAV1-YAP/TAZ fine-tune the fibrotic response through the modulation of MMT, onto which TGF-ß1-dependent signaling coordinately converges. Our findings reveal a cooperation between biomechanical and biochemical signals in the triggering of MMT, representing a novel potential opportunity to intervene mechanically induced disorders coursing with peritoneal fibrosis, such as post-surgical adhesions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Caveolin 1/metabolism , Peritoneal Fibrosis/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/physiology , Animals , Caveolin 1/physiology , Caveolins/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Peritoneal Dialysis/methods , Peritoneal Fibrosis/genetics , Peritoneal Fibrosis/pathology , Peritoneum/metabolism , Signal Transduction/drug effects , Smad3 Protein/metabolism , Tissue Adhesions/metabolism , Transcription Factors/physiology , Transforming Growth Factor beta1/metabolism , YAP-Signaling Proteins
7.
Cancer Metastasis Rev ; 39(2): 485-503, 2020 06.
Article in English | MEDLINE | ID: mdl-32514892

ABSTRACT

Tumor stiffening is a hallmark of malignancy that actively drives tumor progression and aggressiveness. Recent research has shed light onto several molecular underpinnings of this biomechanical process, which has a reciprocal crosstalk between tumor cells, stromal fibroblasts, and extracellular matrix remodeling at its core. This dynamic communication shapes the tumor microenvironment; significantly determines disease features including therapeutic resistance, relapse, or metastasis; and potentially holds the key for novel antitumor strategies. Caveolae and their components emerge as integrators of different aspects of cell function, mechanotransduction, and ECM-cell interaction. Here, we review our current knowledge on the several pivotal roles of the essential caveolar component caveolin-1 in this multidirectional biomechanical crosstalk and highlight standing questions in the field.


Subject(s)
Caveolin 1/metabolism , Neoplasms/metabolism , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Communication/physiology , Disease Progression , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Mechanotransduction, Cellular , Neoplasms/pathology , Receptor Cross-Talk , Stromal Cells/metabolism , Stromal Cells/pathology
8.
Prog Mol Subcell Biol ; 57: 203-234, 2018.
Article in English | MEDLINE | ID: mdl-30097777

ABSTRACT

Caveolae are 60-80 nm invaginated plasma membrane (PM) nanodomains, with a specific lipid and protein composition, which assist and regulate multiple processes in the plasma membrane-ranging from the organization of signalling complexes to the mechanical adaptation to changes in PM tension. However, since their initial descriptions, these structures have additionally been found tightly linked to internalization processes, mechanoadaptation, to the regulation of signalling events and of endosomal trafficking. Here, we review caveolae biology from this perspective, and its implications for cell physiology and disease.


Subject(s)
Cell Membrane/genetics , Endocytosis/genetics , Metabolic Networks and Pathways/genetics , Protein Transport/genetics , Animals , Caveolae/metabolism , Cell Membrane/metabolism , Humans , Signal Transduction/genetics
9.
Sci Rep ; 8(1): 6476, 2018 Apr 19.
Article in English | MEDLINE | ID: mdl-29674750

ABSTRACT

A correction to this article has been published and is linked from the HTML version of this paper. The error has not been fixed in the paper.

10.
Sci Rep ; 7(1): 16497, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29184100

ABSTRACT

Inositol Requiring Enzyme-1 (IRE1) is the most conserved transducer of the Unfolded Protein Response (UPR), a surveillance mechanism that ensures homeostasis of the endoplasmic reticulum (ER) in eukaryotes. IRE1 activation orchestrates adaptive responses, including lipid anabolism, metabolic reprogramming, increases in protein folding competency, and ER expansion/remodeling. However, we still know surprisingly little regarding the principles by which this ER transducer is deactivated upon ER stress clearance. Here we show that Protein Kinase B-mechanistic Target of Rapamycin (PKB/AKT-mTOR) signaling controls the dynamics of IRE1 deactivation by regulating ER-mitochondria physical contacts and the autophosphorylation state of IRE1. AKT-mTOR-mediated attenuation of IRE1 activity is important for ER remodelling dynamics and cell survival in the face of recursive, transient ER stress. Our observations suggest that IRE1 attenuation is an integral component of anabolic programmes regulated by AKT-mTOR. We suggest that AKT-mTOR activity is part of a 'timing mechanism' to deactivate IRE1 immediately following engagement of the UPR, in order to limit prolonged IRE1 RNAse activity that could lead to damaging inflammation or apoptosis.

11.
Methods Mol Biol ; 1487: 163-174, 2017.
Article in English | MEDLINE | ID: mdl-27924566

ABSTRACT

Caveolin1, the main component of caveolae, plays a major role in regulating cell motility, gene expression, and cytoskeleton remodeling downstream of many membrane receptors. Here, we summarize different techniques set up to study changes in cell morphology and cell motility regulated by ERK/caveolin1 interactions during induction of epithelial mesenchymal transition (EMT) in mesothelial cells (MCs).


Subject(s)
Caveolin 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Signal Transduction , Animals , Cell Culture Techniques , Cell Movement , Cell Shape , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Mice , Peritoneal Cavity , Protein Binding
12.
Cell Chem Biol ; 23(11): 1372-1382, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27746127

ABSTRACT

Embryonic stem cells (ESC) have the potential to generate all the cell lineages that form the body. However, the molecular mechanisms underlying ESC differentiation and especially the role of alternative splicing in this process remain poorly understood. Here, we show that the alternative splicing regulator MBNL1 promotes generation of the atypical calcineurin Aß variant CnAß1 in mouse ESCs (mESC). CnAß1 has a unique C-terminal domain that drives its localization mainly to the Golgi apparatus by interacting with Cog8. CnAß1 regulates the intracellular localization and activation of the mTORC2 complex. CnAß1 knockdown results in delocalization of mTORC2 from the membrane to the cytoplasm, inactivation of the AKT/GSK3ß/ß-catenin signaling pathway, and defective mesoderm specification. In summary, here we unveil the structural basis for the mechanism of action of CnAß1 and its role in the differentiation of mESCs to the mesodermal lineage.


Subject(s)
Calcineurin/metabolism , Mouse Embryonic Stem Cells/cytology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Calcineurin/analysis , Cell Differentiation , Cell Line , Golgi Apparatus/metabolism , Mechanistic Target of Rapamycin Complex 2 , Mice , Mouse Embryonic Stem Cells/metabolism , Multiprotein Complexes/analysis , Signal Transduction , TOR Serine-Threonine Kinases/analysis
13.
Stem Cells Int ; 2016: 3543678, 2016.
Article in English | MEDLINE | ID: mdl-26941801

ABSTRACT

Peritoneal dialysis is a form of renal replacement alternative to the hemodialysis. During this treatment, the peritoneal membrane acts as a permeable barrier for exchange of solutes and water. Continual exposure to dialysis solutions, as well as episodes of peritonitis and hemoperitoneum, can cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy, eventually leading to discontinuation of the peritoneal dialysis. Among the different events controlling this pathological process, epithelial to mesenchymal transition of mesothelial cells plays a main role in the induction of fibrosis and in subsequent functional deterioration of the peritoneal membrane. Here, the main extracellular inducers and cellular players are described. Moreover, signaling pathways acting during this process are elucidated, with emphasis on signals delivered by TGF-ß family members and by Toll-like/IL-1ß receptors. The understanding of molecular mechanisms underlying fibrosis of the peritoneal membrane has both a basic and a translational relevance, since it may be useful for setup of therapies aimed at counteracting the deterioration as well as restoring the homeostasis of the peritoneal membrane.

14.
Blood ; 127(15): 1930-9, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26837700

ABSTRACT

Caveolin-1 (Cav-1) is a key organizer of membrane specializations and a scaffold protein that regulates signaling in multiple cell types. We found increased Cav-1 expression in human and murine T cells after allogeneic hematopoietic cell transplantation. Indeed, Cav-1(-/-)donor T cells caused less severe acute graft-versus-host disease (GVHD) and yielded higher numbers of regulatory T cells (Tregs) compared with controls. Depletion of Tregs from the graft abrogated this protective effect. Correspondingly, Treg frequencies increased when Cav-1(-/-)T cells were exposed to transforming growth factor-ß/T-cell receptor (TCR)/CD28 activation or alloantigen stimulation in vitro compared with wild-type T cells. Mechanistically, we found that the phosphorylation of Cav-1 is dispensable for the control of T-cell fate by using a nonphosphorylatable Cav-1 (Y14F/Y14F) point-mutation variant. Moreover, the close proximity of lymphocyte-specific protein tyrosine kinase (Lck) to the TCR induced by TCR-activation was reduced in Cav-1(-/-)T cells. Therefore, less TCR/Lck clustering results in suboptimal activation of the downstream signaling events, which correlates with the preferential development into a Treg phenotype. Overall, we report a novel role for Cav-1 in TCR/Lck spatial distribution upon TCR triggering, which controls T-cell fate toward a regulatory phenotype. This alteration translated into a significant increase in the frequency of Tregs and reduced GVHD in vivo.


Subject(s)
Caveolin 1/metabolism , Caveolin 1/physiology , Gene Expression Regulation , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , Adaptor Proteins, Signal Transducing/genetics , Animals , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , Caveolin 1/genetics , Cell Differentiation , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphorylation , Prospective Studies , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta/metabolism , Transplantation, Homologous
15.
Mol Cell Proteomics ; 15(5): 1740-60, 2016 05.
Article in English | MEDLINE | ID: mdl-26893027

ABSTRACT

The coordinated behavior of proteins is central to systems biology. However, the underlying mechanisms are poorly known and methods to analyze coordination by conventional quantitative proteomics are still lacking. We present the Systems Biology Triangle (SBT), a new algorithm that allows the study of protein coordination by pairwise quantitative proteomics. The Systems Biology Triangle detected statistically significant coordination in diverse biological models of very different nature and subjected to different kinds of perturbations. The Systems Biology Triangle also revealed with unprecedented molecular detail an array of coordinated, early protein responses in vascular smooth muscle cells treated at different times with angiotensin-II. These responses included activation of protein synthesis, folding, turnover, and muscle contraction - consistent with a differentiated phenotype-as well as the induction of migration and the repression of cell proliferation and secretion. Remarkably, the majority of the altered functional categories were protein complexes, interaction networks, or metabolic pathways. These changes could not be detected by other algorithms widely used by the proteomics community, and the vast majority of proteins involved have not been described before to be regulated by AngII. The unique capabilities of The Systems Biology Triangle to detect functional protein alterations produced by the coordinated action of proteins in pairwise quantitative proteomics experiments make this algorithm an attractive choice for the biological interpretation of results on a routine basis.


Subject(s)
Proteome/analysis , Proteomics/methods , Systems Biology/methods , Algorithms , Animals , High-Throughput Screening Assays , Humans , Protein Interaction Maps
16.
Nat Commun ; 6: 7292, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26073653

ABSTRACT

Biological processes in any physiological environment involve changes in cell shape, which must be accommodated by their physical envelope--the bilayer membrane. However, the fundamental biophysical principles by which the cell membrane allows for and responds to shape changes remain unclear. Here we show that the 3D remodelling of the membrane in response to a broad diversity of physiological perturbations can be explained by a purely mechanical process. This process is passive, local, almost instantaneous, before any active remodelling and generates different types of membrane invaginations that can repeatedly store and release large fractions of the cell membrane. We further demonstrate that the shape of those invaginations is determined by the minimum elastic and adhesive energy required to store both membrane area and liquid volume at the cell-substrate interface. Once formed, cells reabsorb the invaginations through an active process with duration of the order of minutes.


Subject(s)
Adaptation, Physiological/physiology , Cell Membrane/physiology , Fibroblasts/physiology , Animals , Cell Shape , Cell Size , Elasticity , Mice , Models, Biological , Models, Theoretical , Osmolar Concentration , Stress, Mechanical
18.
EMBO Mol Med ; 7(1): 102-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25550395

ABSTRACT

Peritoneal dialysis (PD) is a form of renal replacement therapy whose repeated use can alter dialytic function through induction of epithelial-mesenchymal transition (EMT) and fibrosis, eventually leading to PD discontinuation. The peritoneum from Cav1-/- mice showed increased EMT, thickness, and fibrosis. Exposure of Cav1-/- mice to PD fluids further increased peritoneal membrane thickness, altered permeability, and increased the number of FSP-1/cytokeratin-positive cells invading the sub-mesothelial stroma. High-throughput quantitative proteomics revealed increased abundance of collagens, FN, and laminin, as well as proteins related to TGF-ß activity in matrices derived from Cav1-/- cells. Lack of Cav1 was associated with hyperactivation of a MEK-ERK1/2-Snail-1 pathway that regulated the Smad2-3/Smad1-5-8 balance. Pharmacological blockade of MEK rescued E-cadherin and ZO-1 inter-cellular junction localization, reduced fibrosis, and restored peritoneal function in Cav1-/- mice. Moreover, treatment of human PD-patient-derived MCs with drugs increasing Cav1 levels, as well as ectopic Cav1 expression, induced re-acquisition of epithelial features. This study demonstrates a pivotal role of Cav1 in the balance of epithelial versus mesenchymal state and suggests targets for the prevention of fibrosis during PD.


Subject(s)
Caveolin 1/deficiency , Epithelial-Mesenchymal Transition , Peritoneal Dialysis/adverse effects , Peritoneum/physiopathology , Transcription Factors/metabolism , Animals , Caveolin 1/genetics , Epithelial Cells/metabolism , Fibrosis , Humans , MAP Kinase Signaling System , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Peritoneum/enzymology , Peritoneum/metabolism , Peritoneum/pathology , Smad Proteins/genetics , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
19.
PLoS One ; 7(2): e31492, 2012.
Article in English | MEDLINE | ID: mdl-22384029

ABSTRACT

Peritoneal fibrosis is a frequent complication of peritoneal dialysis following repeated low grade inflammatory and pro-fibrotic insults. This pathological process may lead to ultrafiltration failure and eventually to the discontinuing of the therapy. Fibrosis is linked to epithelial to mesenchymal transition (EMT) of the peritoneal mesothelial cells, which acquire invasive and fibrogenic abilities. Here, we analyzed the role of the transforming growth factor-activated kinase-1 (TAK1) in the EMT of primary mesothelial cells from human peritoneum. The inhibition of TAK1 in mesenchymal-like mesothelial cells from the effluents of patients undergoing peritoneal dialysis led to the reacquisition of the apical to basolateral polarity, to increased expression of epithelial and to down-regulation of mesenchymal markers. TAK1 inhibition also resulted in decreased migratory/invasive abilities of effluent-derived mesothelial cells. Simultaneous inhibition of ERK1/2 and TAK1 pathways did not lead to an additive effect in the reacquisition of the epithelial phenotype. Inhibition of TAK1 also blocked EMT in vitro and reduced the levels of PAI-1, which is involved in fibrosis and invasion. Analysis of signalling pathways downstream of TAK1 involved in EMT induction, showed that TAK1 inhibition reduced the transcriptional activity of NF-κB and Smad3, as well as the phosphorylation of c-jun, while enhancing Smad1-5-8 activity. These results demonstrate that TAK1 is a cross-point in a network including different pro-EMT transcription factors, such as NF-κB, Snail, AP-1 and Smads. The identification of TAK1 as a main biochemical mediator of EMT and fibrosis in mesothelial cells from human peritoneum and the study of signalling pathways induced by its activity may be relevant in the design of new therapies aimed to counteract peritoneal fibrosis.


Subject(s)
Gene Expression Regulation , MAP Kinase Kinase Kinases/genetics , Peritoneal Fibrosis/therapy , Down-Regulation , Epithelial-Mesenchymal Transition , Fibrosis , Humans , Interleukin-1beta/metabolism , Mesoderm/pathology , Models, Biological , NF-kappa B/metabolism , Peritoneal Dialysis/methods , Peritoneal Fibrosis/pathology , Phenotype , Phosphorylation , Signal Transduction , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...