Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 14(2): 110, 2023 02 11.
Article in English | MEDLINE | ID: mdl-36774344

ABSTRACT

Mitochondria preserve metabolic homeostasis and integrate stress signals, to trigger cytoprotective, or cell death pathways. Mitochondrial homeostasis and function decline with age. The mechanisms underlying the deterioration of mitochondrial homeostasis during ageing, or in age-associated pathologies, remain unclear. Here, we show that CISD-1, a mitochondrial iron-sulfur cluster binding protein, implicated in the pathogenesis of Wolfram neurodegenerative syndrome type 2, modulates longevity in the nematode Caenorhabditis elegans by engaging autophagy and the mitochondrial intrinsic apoptosis pathway. The anti-apoptotic protein CED-9 is the downstream effector that mediates CISD-1-dependent effects on proteostasis, neuronal integrity and lifespan. Moreover, intracellular iron abundance is critical for CISD-1 function, since mild iron supplementation is sufficient to decelerate ageing and partly ameliorate the disturbed mitochondrial bioenergetics and proteostasis of CISD-1 deficient animals. Our findings reveal that CISD-1 serves as a mechanistic link between autophagy and the apoptotic pathway in mitochondria to differentially modulate organismal proteostasis and ageing, and suggest novel approaches which could facilitate the treatment of Wolfram Syndrome or related diseases.


Subject(s)
Aging , Autophagy , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Proteostasis , Animals , Aging/metabolism , Apoptosis , Autophagy/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Longevity , Mitochondria/metabolism
2.
Cell Adh Migr ; 12(1): 69-85, 2018 01 02.
Article in English | MEDLINE | ID: mdl-28524745

ABSTRACT

Vascular smooth muscle cell (SMC) switching between differentiated and dedifferentiated phenotypes is reversible and accompanied by morphological and functional alterations that require reconfiguration of cell-cell and cell-matrix adhesion networks. Studies attempting to explore changes in overall composition of the adhesion nexus during SMC phenotype transition are lacking. We have previously demonstrated that T-cadherin knockdown enforces SMC differentiation, whereas T-cadherin upregulation promotes SMC dedifferentiation. This study used human aortic SMCs ectopically modified with respect to T-cadherin expression to characterize phenotype-associated cell-matrix adhesion molecule expression, focal adhesions configuration and migration modes. Compared with dedifferentiated/migratory SMCs (expressing T-cadherin), the differentiated/contractile SMCs (T-cadherin-deficient) exhibited increased adhesion to several extracellular matrix substrata, decreased expression of several integrins, matrix metalloproteinases and collagens, and also distinct focal adhesion, adherens junction and intracellular tension network configurations. Differentiated and dedifferentiated phenotypes displayed distinct migrational velocity and directional persistence. The restricted migration efficiency of the differentiated phenotype was fully overcome by reducing actin polymerization with ROCK inhibitor Y-27632 whereas myosin II inhibitor blebbistatin was less effective. Migration efficiency of the dedifferentiated phenotype was diminished by promoting actin polymerization with lysophosphatidic acid. These findings held true in both 2D-monolayer and 3D-spheroid migration models. Thus, our data suggest that despite global differences in the cell adhesion nexus of the differentiated and dedifferentiated phenotypes, structural actin cytoskeleton characteristics per se play a crucial role in permissive regulation of cell-matrix adhesive interactions and cell migration behavior during T-cadherin-induced SMC phenotype transition.


Subject(s)
Actin Cytoskeleton/metabolism , Cadherins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Actin Cytoskeleton/drug effects , Amides/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/drug effects , Cell Communication/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Humans , Integrins/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Pyridines/pharmacology
3.
Sci Rep ; 7(1): 5115, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698593

ABSTRACT

The metabolic and endocrine functions of adipose tissue and the ability of organisms to cope with cellular stress have a direct impact on physiological ageing and the aetiology of various diseases such as obesity-related pathologies and cancer. The endocrine effects of adipose tissue are mediated by secreted adipokines, which modulate metabolic processes and influence related maladies. Although a plethora of molecules and signaling pathways associate ageing with proteotoxic stress and cellular metabolism, our understanding of how these pathways interconnect to coordinate organismal physiology remains limited. We dissected the mechanisms linking adiponectin signalling pathways and endoplasmic reticulum (ER) proteotoxic stress responses that individually or synergistically affect longevity in C. elegans. Animals deficient for the adiponectin receptor PAQR-1 respond to ER stress, by rapidly activating the canonical ER unfolded protein response (UPRER) pathway, which is primed in these animals under physiological conditions by specific stress defence transcription factors. PAQR-1 loss enhances survival and promotes longevity under ER stress and reduced insulin/IGF-1 signalling. PAQR-1 engages UPRER, autophagy and lipase activity to modulate lipid metabolism during ageing. Our findings demonstrate that moderating adiponectin receptor -1 activity extends lifespan under stress, and directly implicate adiponectin signalling as a coupler between proteostasis and lipid metabolism during ageing.


Subject(s)
Adiponectin/metabolism , Aging/metabolism , Caenorhabditis elegans/physiology , Receptors, Adiponectin/metabolism , Animals , Caenorhabditis elegans Proteins/metabolism , Endoplasmic Reticulum Stress , Insulin-Like Growth Factor I/metabolism , Lipid Metabolism , Proteostasis , Signal Transduction , Unfolded Protein Response
4.
Cell Signal ; 35: 163-175, 2017 07.
Article in English | MEDLINE | ID: mdl-28392425

ABSTRACT

Autophagy is an evolutionary conserved intracellular catabolic process of vital importance to cell and tissue homeostasis. Autophagy is implicated in the pathogenesis of atherosclerosis but participating cells, molecular mechanisms and functional outcomes have not been fully elucidated. T-cadherin, an atypical glycosylphosphatidylinositol-anchored member of the cadherin superfamily of adhesion molecules, is upregulated on smooth muscle cells (SMCs)1 in atherosclerotic lesions. Here, using rat and murine aortic SMCs as experimental models, we surveyed the ability of T-cadherin to regulate autophagy in SMCs during serum-starvation stress. Ectopic upregulation of T-cadherin in SMCs resulted in augmented autophagy characterized by increased autophagic flux, LC3-II abundance and autophagosome formation. Analysis of signal transduction pathway effectors and use of specific pharmacological inhibitors demonstrated that T-cadherin-associated enhancement of the autophagic response to serum-deprivation was dependent on MEK1/2/Erk1/2 activation and independent of PI3K/Akt/mTORC1, reactive oxygen species or endoplasmic reticulum stress. T-cadherin upregulation on SMCs conferred a survival advantage during prolonged serum-starvation which was sensitive to inhibition of MEK1/2/Erk1/2 by PD98059 or UO126 and to blockade of autophagy by chloroquine. Loss of T-cadherin expression in SMCs diminished autophagy responsiveness and compromised survival under conditions of serum-starvation. Overall our findings have identified T-cadherin as a novel positive regulator of autophagy and survival in SMCs.


Subject(s)
Autophagy/genetics , Cadherins/genetics , Endoplasmic Reticulum Stress/genetics , Muscle, Smooth, Vascular/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Apoptosis/genetics , Flavonoids/administration & dosage , Gene Expression Regulation/drug effects , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , MAP Kinase Signaling System/drug effects , Mice , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Reactive Oxygen Species/metabolism , Transcriptional Activation/genetics
5.
Cell Stress Chaperones ; 22(4): 481-492, 2017 07.
Article in English | MEDLINE | ID: mdl-28074336

ABSTRACT

Small heat shock proteins (sHSPs) are gatekeepers of cellular homeostasis across species, preserving proteome integrity under stressful conditions. Nonetheless, recent evidence suggests that sHSPs are more than molecular chaperones with merely auxiliary role. In contrast, sHSPs have emerged as central lifespan determinants, and their malfunction has been associated with the manifestation of neurological disorders, cardiovascular disease and cancer malignancies. In this review, we focus on the role of sHSPs in ageing and age-associated diseases and highlight the most prominent paradigms, where impairment of sHSP function has been implicated in human pathology.


Subject(s)
Aging , Heat-Shock Proteins, Small/metabolism , Animals , Cardiovascular Diseases/metabolism , Heat-Shock Proteins, Small/analysis , Humans , Inflammation/metabolism , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Protein Processing, Post-Translational
6.
Methods Mol Biol ; 1292: 215-34, 2015.
Article in English | MEDLINE | ID: mdl-25804759

ABSTRACT

The ageing process is characterized by deterioration of physiological function accompanied by frailty and ageing-associated diseases. The most broadly and well-studied pathways influencing ageing are the insulin/insulin-like growth factor 1 signaling pathway and the dietary restriction pathway. Recent studies in diverse organisms have also delineated emerging pathways, which collectively or independently contribute to ageing. Among them the proteostatic-stress-response networks, inextricably affect normal ageing by maintaining or restoring protein homeostasis to preserve proper cellular and organismal function. In this chapter, we survey the involvement of heat stress and endoplasmic reticulum stress responses in the regulation of longevity, placing emphasis on the cross talk between different response mechanisms and their systemic effects. We further discuss novel insights relevant to the molecular pathways mediating these stress responses that may facilitate the development of innovative interventions targeting age-related pathologies such as diabetes, cancer, cardiovascular and neurodegenerative diseases.


Subject(s)
Aging/physiology , Aging/genetics , Aging/metabolism , Animals , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/physiology , Homeostasis/genetics , Homeostasis/physiology , Humans , Longevity/genetics , Longevity/physiology
7.
Mol Cell Oncol ; 2(2): e975027, 2015.
Article in English | MEDLINE | ID: mdl-27308424

ABSTRACT

The term cancer describes a group of multifaceted diseases characterized by an intricate pathophysiology. Despite significant advances in the fight against cancer, it remains a key public health concern and burden on societies worldwide. Elucidation of key molecular and cellular mechanisms of oncogenic diseases will facilitate the development of better intervention strategies to counter or prevent tumor development. In vivo and in vitro models have long been used to delineate distinct biological processes involved in cancer such as apoptosis, proliferation, angiogenesis, invasion, metastasis, genome instability, and metabolism. In this review, we introduce Caenorhabditis elegans as an emerging animal model for systematic dissection of the molecular basis of tumorigenesis, focusing on the well-established processes of apoptosis and autophagy. Additionally, we propose that C. elegans can be used to advance our understanding of cancer progression, such as deregulation of energy metabolism, stem cell reprogramming, and host-microflora interactions.

8.
FASEB J ; 29(2): 494-507, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25381040

ABSTRACT

T-cadherin is an atypical glycosylphosphatidylinsoitol-anchored member of the cadherin superfamily of adhesion molecules. We found that T-cadherin overexpression in malignant (DU145) and benign (BPH-1) prostatic epithelial cell lines or silencing in the BPH-1 cell line, respectively, promoted or inhibited migration and spheroid invasion in collagen I gel and Matrigel. T-cadherin-dependent effects were associated with changes in cell phenotype: overexpression caused cell dissemination and loss of polarity evaluated by relative positioning of the Golgi/nuclei in cell groups, whereas silencing caused formation of compact polarized epithelial-like clusters. Epidermal growth factor receptor (EGFR) and IGF factor-1 receptor (IGF-1R) were identified as mediators of T-cadherin effects. These receptors per se had opposing influences on cell phenotype. EGFR activation with EGF or IGF-1R inhibition with NVP-AEW541 promoted dissemination, invasion, and polarity loss. Conversely, inhibition of EGFR with gefitinib or activation of IGF-1R with IGF-1 rescued epithelial morphology and decreased invasion. T-cadherin silencing enhanced both EGFR and IGF-1R phosphorylation, yet converted cells to the morphology typical for activated IGF-1R. T-cadherin effects were sensitive to modulation of EGFR or IGF-1R activity, suggesting direct involvement of both receptors. We conclude that T-cadherin regulates prostate cancer cell behavior by tuning the balance in EGFR/IGF-1R activity and enhancing the impact of IGF-1R.


Subject(s)
Cadherins/metabolism , ErbB Receptors/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Receptor, IGF Type 1/metabolism , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Cell Survival , Collagen/chemistry , Drug Combinations , Gefitinib , Gene Silencing , Golgi Apparatus/metabolism , Humans , Laminin/chemistry , Male , Neoplasm Invasiveness , Phenotype , Phosphorylation , Proteoglycans/chemistry , Pyrimidines/chemistry , Pyrroles/chemistry , Quinazolines/chemistry
9.
Trends Endocrinol Metab ; 25(4): 212-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24388148

ABSTRACT

Human lifespan has been increasing steadily during modern times, mainly due to medical advancements that combat infant mortality and various life-threatening diseases. However, this gratifying longevity rise is accompanied by growing incidences of devastating age-related pathologies. Understanding the cellular and molecular mechanisms that underlie aging and regulate longevity is of utmost relevance towards offsetting the impact of age-associated disorders and increasing the quality of life for the elderly. Several evolutionarily conserved pathways that modulate lifespan have been identified in organisms ranging from yeast to primates. Here we survey recent findings highlighting the interplay of various genetic, epigenetic, and cell-specific factors, and also symbiotic relationships, as longevity determinants. We further discuss outstanding matters within the framework of emerging, integrative views of aging.


Subject(s)
Aging/physiology , Longevity/physiology , Aging/genetics , Humans , Longevity/genetics , Signal Transduction/genetics , Signal Transduction/physiology
10.
Cell Signal ; 25(5): 1044-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23411345

ABSTRACT

Reciprocal cross-talk between receptor tyrosine kinases (RTKs) and classical cadherins (e.g. EGFR/E-cadherin, VEGFR/VE-cadherin) has gained appreciation as a combinatorial molecular mechanism enabling diversification of the signalling environment and according differential cellular responses. Atypical glycosylphosphatidylinositol (GPI)-anchored T-cadherin (T-cad) was recently demonstrated to function as a negative auxiliary regulator of EGFR pathway activation in A431 squamous cell carcinoma (SCC) cells. Here we investigate the reciprocal impact of EGFR activation on T-cad. In resting A431 T-cad was distributed globally over the cell body. Following EGF stimulation T-cad was redistributed to the sites of cell-cell contact where it colocalized with phosphorylated EGFR(Tyr1068). T-cad redistribution was not affected by endomembrane protein trafficking inhibitor brefeldin A or de novo protein synthesis inhibitor cycloheximide, supporting mobilization of plasma membrane associated T-cad. EGF-induced relocalization of T-cad to cell-cell contacts could be abrogated by specific inhibitors of EGFR tyrosine kinase activity (gefitinib or lapatinib), lipid raft integrity (filipin), actin microfilament polymerization (cytochalasin D or cytochalasin B), p38MAPK (SB203580) or Rac1 (compound4). Erk1/2 inhibitor PD98059 increased phospho-EGFR(tyr1068) levels and not only amplified effects of EGF but also per se promoted some relocalization of T-cad to cell-cell contacts. Rac1 activation by EGF was inhibited by gefitinib, lapatinib or SB203580 but amplified by PD98059. Taken together our data suggest that T-cad translocation to cell-cell contacts is sensitive to the activity status of EGFR, requires lipid raft domain integrity and actin filament polymerization, and crucial intracellular signalling mediators include Rac1 and p38MAPK. The study has revealed a novel aspect of reciprocal cross-talk between EGFR and T-cad.


Subject(s)
Cadherins/metabolism , ErbB Receptors/metabolism , Cell Communication/drug effects , Cell Line, Tumor , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Lapatinib , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
11.
Eur J Cancer ; 49(8): 2048-58, 2013 May.
Article in English | MEDLINE | ID: mdl-23369463

ABSTRACT

T-cadherin is gaining recognition as a determinant for the development of incipient invasive squamous cell carcinoma (SCC). However, effects of T-cadherin expression on the metastatic potential of SCC have not been studied. Here, using a murine model of experimental metastasis following tail vein injection of A431 SCC cells we report that loss of T-cadherin increased both the incidence and rate of appearance of lung metastases. T-cadherin-silenced SCC metastases were highly disordered with evidence of single cell dissemination away from main foci whereas SCC metastases overexpressing T-cadherin developed as compact, tightly organised sheets. SCC cell adhesion to vascular endothelial cells (EC) in culture was increased for T-cadherin-silenced SCC and decreased for T-cadherin-overexpressing SCC. Confocal microscopy showed that T-cadherin-silenced SCC adherent on EC display an elongated morphology with long thin extensions and a high degree of intercalation within the EC monolayer, whereas SCC overexpressing T-cadherin formed poorly-spread multicellular aggregates that remain on the outer surface of the EC monolayer. T-cadherin-deficient SCC or human keratinocyte cells exhibited increased transendothelial migration in vitro which could be attenuated in the presence of EGFR inhibitor gefitinib. Our data suggest that loss of T-cadherin can increase metastatic potential and aggressiveness of SCC, possibly due to facilitating arrest and extravasation through the vascular wall and/or more efficient establishment of metastases in the new microenvironment.


Subject(s)
Cadherins/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Skin Neoplasms/metabolism , Animals , Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Adhesion/genetics , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gefitinib , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoblotting , Keratinocytes/cytology , Keratinocytes/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Confocal , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , RNA Interference , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/genetics , Transplantation, Heterologous
12.
J Invest Dermatol ; 132(9): 2275-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22592160

ABSTRACT

Genetic and epigenetic studies in different cancers, including cutaneous carcinomas, have implicated T-cadherin (T-cad) as a tumor suppressor. Immunohistochemical and in vitro studies have suggested that T-cad loss promotes incipient invasiveness in cutaneous squamous cell carcinoma (SCC). Molecular mechanisms are unknown. This study found that the main consequence of T-cad silencing in SCC is facilitation of ligand-dependent EGFR activation, whereas T-cad overexpression impedes EGFR activation. Gain- and loss-of-function studies in A431 SCC cells demonstrate T-cad-controlled responsiveness to EGF with respect to pharmacological inhibition of EGFR and to diverse signaling and functional events of the EGFR activation cascade (EGFR phosphorylation, internalization, nuclear translocation, cell retraction/de-adhesion, motility, invasion, integrin ß1, and Rho small GTPases such as RhoA, Rac1, and Cdc42 activation). Further, T-cad modulates the EGFR pathway activity by influencing membrane compartmentalization of EGFR; T-cad upregulation promotes retention of EGFR in lipid rafts, whereas T-cad silencing releases EGFR from this compartment, rendering EGFR more accessible to ligand stimulation. This study reveals a mechanism for fine-tuning of EGFR activity in SCC, whereby T-cad represents an auxiliary "negative" regulator of the EGFR pathway, which impacts invasion-associated behavioral responses of SCC to EGF. This action of T-cad in SCC may serve as a paradigm explaining other malignancies displaying concomitant T-cad loss and enhanced EGFR activity.


Subject(s)
Cadherins/physiology , Carcinoma, Squamous Cell/pathology , Cell Movement , ErbB Receptors/physiology , Signal Transduction/physiology , Skin Neoplasms/pathology , Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Gefitinib , Gene Silencing , Humans , Lapatinib , Membrane Microdomains/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Skin Neoplasms/genetics , rho GTP-Binding Proteins/metabolism
13.
Cardiovasc Res ; 93(3): 498-507, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22235028

ABSTRACT

AIMS: T-cadherin (T-cad) is a glycosylphosphatidylinositol-anchored cadherin family member. Experimental, clinical, and genomic studies suggest a role for T-cad in vascular disorders such as atherosclerosis and hypertension, which are associated with endothelial dysfunction and insulin resistance (InsRes). In endothelial cells (EC), T-cad and insulin activate similar signalling pathways [e.g. PI3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR)] and processes (e.g. angiogenesis). We hypothesize that T-cad is a regulatory component of insulin signalling in EC and therefore a determinant of the development of endothelial InsRes. METHODS AND RESULTS: We investigated T-cad-dependent effects on insulin sensitivity using human EC stably transduced with respect to T-cad overexpression or T-cad silencing. Responsiveness to insulin was examined at the level of effectors of the insulin signalling cascade, EC nitric oxide synthase (eNOS) activation, and angiogenic behaviour. Overexpression and ligation of T-cad on EC attenuates insulin-dependent activation of the PI3K/Akt/mTOR signalling axis, eNOS, EC migration, and angiogenesis. Conversely, T-cad silencing enhances these actions of insulin. Attenuation of EC responsiveness to insulin results from T-cad-mediated chronic activation of the Akt/mTOR-dependent negative feedback loop of the insulin cascade and enhanced degradation of the insulin receptor (IR) substrate. Co-immunoprecipitation experiments revealed an association between T-cad and IR. Filipin abrogated inhibitory effects of T-cad on insulin signalling, demonstrating localization of T-cad-insulin cross-talk to lipid raft plasma membrane domains. Hyperinsulinaemia up-regulates T-cad mRNA and protein levels in EC. CONCLUSION: T-cad expression modulates signalling and functional responses of EC to insulin. We have identified a novel signalling mechanism regulating insulin function in the endothelium and attribute a role for T-cad up-regulation in the pathogenesis of endothelial InsRes.


Subject(s)
Cadherins/metabolism , Endothelial Cells/metabolism , Insulin/metabolism , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/metabolism , Signal Transduction/physiology , Cadherins/genetics , Cell Line , Endothelial Cells/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Gene Silencing , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/physiology , Humans , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Membrane Microdomains/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptor, Insulin/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
J Leukoc Biol ; 90(5): 929-39, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21807744

ABSTRACT

iNKT cells are a unique T cell subset, which is CD1d-restricted and specific for glycolipid antigens. In advanced atherosclerotic plaques, focal collections of inflammatory cells correlate with areas of intraplaque neovascularization. We reported recently that iNKT cells might facilitate intraplaque neovascularization by enhancing EC migration and sprouting in an IL-8-dependent manner. This study investigated the participating effector mechanisms. In ECs, CM, derived from antigen-stimulated human iNKT cells (CM+), induced up-regulation of IL-8R CXCR2 and the phosphorylation of EGFR and of multiple intracellular signaling effectors, including FAK, Src, Erk, Jnk, p38-MAPK, and STAT1 and -3. We found that a cascade of events, which were IL-8-dependent and involved EGFR activation, was responsible for signaling through FAK and Src kinases and necessary for acquisition of angiogenic morphology, migration in a two-dimensional wound assay, and sprout outgrowth in a three-dimensional model of angiogenesis in vitro. The data support that IL-8-dependent activation of angiogenic behavior in ECs, in response to activated iNKT, involves CXCR2, transactivation of EGFR, and subsequent FAK/Src signaling. We found too that activated iNKT increased VEGFR2 expression in ECs. Functional studies confirmed that EGF is the motogenic-enhancing factor in CM+ and is necessary, together with an exogenous source of VEGF, for iNKT-promoted sprout formation. EGFR inhibition may represent a novel therapeutic modality aimed at plaque stabilization through control of neovascularization within developing atherosclerotic plaques.


Subject(s)
Endothelial Cells/immunology , ErbB Receptors/immunology , Interleukin-8/immunology , Lymphocyte Activation/physiology , Natural Killer T-Cells/immunology , Neovascularization, Pathologic , Antigens/immunology , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Communication , Cell Line , Cell Movement , Endothelial Cells/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Humans , Inflammation , Interleukin-8/metabolism , Lipids/immunology , Natural Killer T-Cells/metabolism , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Phosphorylation , Receptors, Interleukin-8B/immunology , Receptors, Interleukin-8B/metabolism , Signal Transduction , Up-Regulation
15.
J Pathol ; 225(4): 512-24, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21766307

ABSTRACT

Mechanisms underlying cutaneous squamous cell carcinoma (SCC) tumour growth and invasion are incompletely understood. Our previous pathological and in vitro studies suggest that cell surface glycoprotein T-cadherin (T-cad) might be a controlling determinant of the behaviour of SCC. Here we used a murine xenograft model to determine whether T-cad modulates SCC tumour progression in vivo. Silencing or up-regulation of T-cad in A431 (shTcad or Tcad(+) , respectively) both resulted in increased tumour expansion in vivo. To explain this unanticipated outcome, we focused on proliferation, apoptosis and angiogenesis/lymphangiogenesis, which are important determinants of the progression of solid tumours in vivo. shTcad exhibited enhanced proliferation potential in vitro and in vivo, and their signalling response to EGF was characterized by a higher Erk1/2:p38MAPK activity ratio, which has been correlated with more aggressive tumour growth. T-cad over-expression did not affect proliferation but staining for cleaved caspase 3 revealed a minimal occurrence of extensive apoptosis in Tcad(+) tumours. Immunofluoresence staining of xenograft sections revealed increased intra-tumoural total microvessel (CD31(+)) and lymphatic vessel (LYVE-1(+)) densities in Tcad(+) tumours. shTcad tumours exhibited decreased microvessel and lymphatic densities. Tcad(+) expressed higher levels of transcripts for VEGF-A, VEGF-C and VEGF-D in vitro and in vivo. Culture supernatants collected from Tcad(+) enhanced sprout outgrowth from spheroids composed of either microvascular or lymphatic endothelial cells, and these in vitro angiogenic and lymphangiogenic responses were abrogated by inclusion of neutralizing VEGF antibodies. We conclude that T-cad can exert pleiotropic effects on SCC progression; up- or down-regulation of T-cad can promote SCC tumour expansion in vivo but through distinct mechanisms, namely enhancement of angio/lymphangiogenic potential or enhancement of proliferation capacity.


Subject(s)
Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Skin Neoplasms/genetics , Animals , Apoptosis , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Disease Progression , Gene Silencing , Glycoproteins/metabolism , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Membrane Transport Proteins , Mice , Microvessels/drug effects , Microvessels/metabolism , Microvessels/pathology , Neovascularization, Pathologic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
16.
Eur Heart J ; 32(6): 760-71, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20584775

ABSTRACT

AIMS: The presence of endothelial cell (EC)-derived surface molecules in the circulation is among hallmarks of endothelial activation and damage in vivo. Previous investigations suggest that upregulation of T-cadherin (T-cad) on the surface of ECs may be a characteristic marker of EC activation and stress. We investigated whether T-cad might also be shed from ECs and in amounts reflecting the extent of activation or damage. METHODS AND RESULTS: Immunoblotting showed the presence of T-cad protein in the culture medium from normal proliferating ECs and higher levels in the medium from stressed/apoptotic ECs. Release of T-cad into the circulation occurs in vivo and in association with endothelial dysfunction. Sandwich ELISA revealed negligible T-cad protein in the plasma of healthy volunteers (0.90 ± 0.90 ng/mL, n = 30), and increased levels in the plasma from patients with non-significant atherosclerosis (9.23 ± 2.61 ng/mL, n = 63) and patients with chronic coronary artery disease (6.93 ± 1.31 ng/mL, n = 162). In both patient groups there was a significant (P = 0.043) dependency of T-cad and degree of endothelial dysfunction as measured by reactive hyperaemia peripheral tonometry. Flow cytometry analysis showed that the major fraction of T-cad was released into the EC culture medium and the plasma as a surface component of EC-derived annexin V- and CD144/CD31-positive microparticles (MPs). Gain-of-function and loss-of-function studies demonstrate that MP-bound T-cad induced Akt phosphorylation and activated angiogenic behaviour in target ECs via homophilic-based interactions. CONCLUSION: Our findings reveal a novel mechanism of T-cad-dependent signalling in the vascular endothelium. We identify T-cad as an endothelial MP antigen in vivo and demonstrate that its level in plasma is increased in early atherosclerosis and correlates with endothelial dysfunction.


Subject(s)
Cadherins/metabolism , Coronary Artery Disease/diagnosis , Aged , Analysis of Variance , Cell-Derived Microparticles/metabolism , Cells, Cultured , Early Diagnosis , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Signal Transduction , Umbilical Veins/cytology , Umbilical Veins/metabolism
17.
Eur J Immunol ; 40(11): 3268-79, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21061446

ABSTRACT

Atherosclerosis, a chronic inflammatory lipid storage disease of large arteries, is complicated by cardiovascular events usually precipitated by plaque rupture or erosion. Inflammation participates in lesion progression and plaque rupture. Identification of leukocyte populations involved in plaque destabilization is important for effective prevention of cardiovascular events. This study investigates CD1d-expressing cells and invariant NKT cells (iNKT) in human arterial tissue, their correlation with disease severity and symptoms, and potential mechanisms for their involvement in plaque formation and/or destabilization. CD1d-expressing cells were present in advanced plaques in patients who suffered from cardiovascular events in the past and were most abundant in plaques with ectopic neovascularization. Confocal microscopy detected iNKT cells in plaques, and plaque-derived iNKT cell lines promptly produced proinflammatory cytokines when stimulated by CD1d-expressing APC-presenting α-galactosylceramide lipid antigen. Furthermore, iNKT cells were diminished in the circulating blood of patients with symptomatic atherosclerosis. Activated iNKT cell-derived culture supernatants showed angiogenic activity in a human microvascular endothelial cell line HMEC-1-spheroid model of in vitro angiogenesis and strongly activated human microvascular endothelial cell line HMEC-1 migration. This functional activity was ascribed to IL-8 released by iNKT cells upon lipid recognition. These findings introduce iNKT cells as novel cellular candidates promoting plaque neovascularization and destabilization in human atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Cell Movement/immunology , Endothelial Cells/immunology , Natural Killer T-Cells/immunology , Neovascularization, Pathologic/immunology , Adult , Aged , Aged, 80 and over , Antigens, CD1d/biosynthesis , Antigens, CD1d/immunology , Arteries/immunology , Arteries/metabolism , Arteries/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Line , Cytokines/biosynthesis , Cytokines/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Male , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
18.
Cell Signal ; 22(9): 1308-16, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20457250

ABSTRACT

Endoplasmic reticulum (ER) stress activated by perturbations in ER homeostasis induces the unfolded protein response (UPR) with chaperon Grp78 as the key activator of UPR signalling. The aim of UPR is to restore normal ER function; however prolonged or severe ER stress triggers apoptosis of damaged cells to ensure protection of the whole organism. Recent findings support an association of ER stress-induced apoptosis of vascular cells with cardiovascular pathologies. T-cadherin (T-cad), an atypical glycosylphosphatidylinositol-anchored member of the cadherin superfamily is upregulated in atherosclerotic lesions. Here we investigate the ability of T-cad to influence UPR signalling and endothelial cell (EC) survival during ER stress. EC were treated with a variety of ER stress-inducing compounds (thapsigargin, dithiothereitol, brefeldin A, tunicamycin, A23187 or homocysteine) and induction of ER stress validated by increases in levels of UPR signalling molecules Grp78 (glucose-regulated protein of 78kDa), phospho-eIF2alpha (phosphorylated eukaryotic initiation factor 2alpha) and CHOP (C/EBP homologous protein). All compounds also increased T-cad mRNA and protein levels. Overexpression or silencing of T-cad in EC respectively attenuated or amplified the ER stress-induced increase in phospho-eIF2alpha, Grp78, CHOP and active caspases. Effects of T-cad-overexpression or T-cad-silencing on ER stress responses in EC were not affected by inclusion of either N-acetylcysteine (reactive oxygen species scavenger), LY294002 (phosphatidylinositol-3-kinase inhibitor) or SP6000125 (Jun N-terminal kinase inhibitor). The data suggest that upregulation of T-cad on EC during ER stress attenuates the activation of the proapoptotic PERK (PKR (double-stranded RNA-activated protein kinase)-like ER kinase) branch of the UPR cascade and thereby protects EC from ER stress-induced apoptosis.


Subject(s)
Apoptosis , Cadherins/metabolism , Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Unfolded Protein Response , Cell Survival , Endoplasmic Reticulum Chaperone BiP , Endothelial Cells/enzymology , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , eIF-2 Kinase/antagonists & inhibitors
19.
FASEB J ; 23(11): 4011-21, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19638398

ABSTRACT

T-cadherin (T-cad) promotes survival, proliferation, and migration of endothelial cells and induces angiogenesis. We aimed to identify domains of T-cad functionally relevant to its effects on endothelial cell behavior. To specifically target the functional properties of the 5 cadherin repeat domains (EC1-EC5) of T-cad, endothelial cells were transduced with lentivectors containing specific T-cad-domain-deletion mutant constructs (DeltaI, DeltaII, DeltaIII, DeltaIV, DeltaV). Empty (E) lentivector-transduced cells served as control. Similarly to overexpression of native T-cad, cells expressing DeltaII, DeltaIII, or DeltaIV displayed elevated levels of p-Akt and p-GSK3beta and increased proliferation rates (for DeltaII, DeltaIII) vs. E. DeltaI- and DeltaV-transduced cells exhibited reduced levels of p-Akt and p-GSK3beta and retarded growth rates vs. E. Stimulatory effects of native T-cad overexpression on Akt and GSK3beta phosphorylation were dose dependently inhibited by coexpression of DeltaI or DeltaV. Subsequent functional analyses compared only DeltaI-, DeltaII-, and DeltaV-mutant constructs with E as a negative control. Unlike DeltaII cells, DeltaI and DeltaV cells failed to exhibit homophilic ligation and deadhesion responses on a substratum of T-cad protein. In the wound assay, migration was increased for DeltaII cells but impaired for DeltaI and DeltaV cells. In endothelial cell-spheroid assay, angiogenic sprouting was augmented for DeltaII cells but inhibited for DeltaI and DeltaV cells. We conclude that EC1 and EC5 domains of T-cad are essential for its proangiogenic effects. DeltaI and DeltaV constructs may serve as dominant-negative mutants and as potential tools targeting excessive angiogenesis.


Subject(s)
Cadherins/chemistry , Cadherins/physiology , Endothelial Cells/physiology , Angiogenesis Inducing Agents , Cadherins/genetics , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Endothelium, Vascular/growth & development , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Humans , Neovascularization, Physiologic/physiology , Protein Structure, Tertiary , Wound Healing/physiology
20.
Cell Signal ; 21(7): 1035-44, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19399994

ABSTRACT

Cadherins are a superfamily of transmembrane proteins that mediate calcium-dependent intercellular adhesion. T-cadherin (T-cad, H-cadherin or cadherin-13) is an atypical member, lacking transmembrane and cytosolic domains and possessing a glycosylphosphatidylinositol moiety that anchors T-cadherin to the plasma membrane. This article reviews current knowledge on the biomolecular characteristics of T-cadherin,its expression and function in different tissues in health and disease and its mechanisms of signal transduction. The structural characteristics of T-cadherin protein predict that it is unlikely to function as a"true" adhesion molecule in vivo. Studies from different fields suggest that it may act rather as a signalling receptor participating in recognition of the environment and regulation of cell motility, proliferation and phenotype. Cellular expression levels of T-cadherin in various tissues frequently correlate (be it negatively or positively) with the proliferative potential of the cells. Loss- and gain-of-function studies demonstrate the ability of T-cadherin to modulate cell motility and growth. Gathering evidence suggests that the "functional predestination" of T-cadherin is in control of tissue architecture through "guiding" navigation of moving structures, segregating functional tissue compartments and "guarding" integrity of functionally connected tissue layers.


Subject(s)
Cadherins/metabolism , Cadherins/chemistry , Cadherins/genetics , Disease , Health , Humans , Protein Binding , Protein Transport , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...