Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(5)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300714

ABSTRACT

TAR DNA-binding protein 43 (TDP-43) is a DNA/RNA-binding protein that regulates gene expression, and its malfunction in neurons has been causally associated with multiple neurodegenerative disorders. Although progress has been made in understanding the functions of TDP-43 in neurons, little is known about its roles in endothelial cells (ECs), angiogenesis, and vascular function. Using inducible EC-specific TDP-43-KO mice, we showed that TDP-43 is required for sprouting angiogenesis, vascular barrier integrity, and blood vessel stability. Postnatal EC-specific deletion of TDP-43 led to retinal hypovascularization due to defects in vessel sprouting associated with reduced EC proliferation and migration. In mature blood vessels, loss of TDP-43 disrupted the blood-brain barrier and triggered vascular degeneration. These vascular defects were associated with an inflammatory response in the CNS with activation of microglia and astrocytes. Mechanistically, deletion of TDP-43 disrupted the fibronectin matrix around sprouting vessels and reduced ß-catenin signaling in ECs. Together, our results indicate that TDP-43 is essential for the formation of a stable and mature vasculature.


Subject(s)
Endothelial Cells , Neuroinflammatory Diseases , Mice , Animals , Endothelial Cells/metabolism , Angiogenesis , Neovascularization, Physiologic/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
2.
Development ; 150(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36852644

ABSTRACT

Wt1 encodes a zinc finger protein that is crucial for epicardium development. Although WT1 is also expressed in coronary endothelial cells (ECs), the abnormal heart development observed in Wt1 knockout mice is mainly attributed to its functions in the epicardium. Here, we have generated an inducible endothelial-specific Wt1 knockout mouse model (Wt1KOΔEC). Deletion of Wt1 in ECs during coronary plexus formation impaired coronary blood vessels and myocardium development. RNA-Seq analysis of coronary ECs from Wt1KOΔEC mice demonstrated that deletion of Wt1 exerted a major impact on the molecular signature of coronary ECs and modified the expression of several genes that are dynamically modulated over the course of coronary EC development. Many of these differentially expressed genes are involved in cell proliferation, migration and differentiation of coronary ECs; consequently, the aforementioned processes were affected in Wt1KOΔEC mice. The requirement of WT1 in coronary ECs goes beyond the initial formation of the coronary plexus, as its later deletion results in defects in coronary artery formation. Through the characterization of these Wt1KOΔEC mouse models, we show that the deletion of Wt1 in ECs disrupts physiological blood vessel formation.


Subject(s)
Coronary Vessels , Endothelial Cells , Mice , Animals , Endothelial Cells/metabolism , Coronary Vessels/metabolism , Pericardium/metabolism , Cell Proliferation/genetics , Neovascularization, Physiologic/genetics , Disease Models, Animal , Mice, Knockout , Myocardium/metabolism , WT1 Proteins/genetics
3.
Int J Mol Sci ; 24(2)2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36674431

ABSTRACT

Synaptic activity is the main energy-consuming process in the central nervous system. We are beginning to understand how energy is supplied and used during synaptic activity by neurons. However, the long-term metabolic adaptations associated with a previous episode of synaptic activity are not well understood. Herein, we show that an episode of synaptic activity increases mitochondrial bioenergetics beyond the duration of the synaptic activity by transcriptionally inducing the expression of iron metabolism genes with the consequent enhancement of cellular and mitochondrial iron uptake. Iron is a necessary component of the electron transport chain complexes, and its chelation or knockdown of mitochondrial iron transporter Mfrn1 blocks the activity-mediated bioenergetics boost. We found that Mfrn1 expression is regulated by the well-known regulator of synaptic plasticity CREB, suggesting the coordinated expression of synaptic plasticity programs with those required to meet the associated increase in energetic demands.


Subject(s)
Energy Metabolism , Neurons , Neurons/metabolism , Mitochondria/metabolism , Biological Transport , Iron/metabolism
4.
Front Cell Dev Biol ; 10: 969100, 2022.
Article in English | MEDLINE | ID: mdl-35874805
5.
PLoS Genet ; 18(6): e1010240, 2022 06.
Article in English | MEDLINE | ID: mdl-35704566

ABSTRACT

Assessing the role of the WT1 transcription factor (WT1) during early gonad differentiation and its impact on adult sex development has been difficult due to the complete gonadal agenesis and embryonic lethality exhibited by Wt1KO mouse models. Here, we generated Wt1LoxP/GFP;Wt1Cre mice, the first Wt1KO mouse model that reaches adulthood with a dramatically reduced Wt1 expression during early gonadogenesis. Wt1LoxP/GFP;Wt1Cre mice lacked mature gonads and displayed genital tracts containing both male and female genital structures and ambiguous genitalia. We found that WT1 is necessary for the activation of both male and female sex-determining pathways, as embryonic mutant gonads failed to upregulate the expression of the genes specific for each genetic programme. The gonads of Wt1LoxP/GFP;Wt1Cre mice showed a lack of production of Sertoli and pre-granulosa cells and a reduced number of germ cells. NR5A1 and the steroidogenic genes expression was modulated differently in XY and XX Wt1LoxP/GFP;Wt1Cre gonads, explaining the mutant phenotypes. Further studies of the XX Wt1LoxP/GFP;Wt1Cre gonads revealed that deletion of WT1 at an early stage impaired the differentiation of several cell types including somatic cells and the ovarian epithelium. Through the characterisation of this Wt1KO mouse model, we show that the deletion of Wt1 during early gonadogenesis produces dramatic defects in adult sex development.


Subject(s)
Gonads , Sex Differentiation , Animals , Cell Differentiation/genetics , Female , Gonads/metabolism , Male , Mice , Ovary/metabolism , Sex Differentiation/genetics , Sexual Development , Testis/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
6.
EMBO Rep ; 22(9): e51954, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34296790

ABSTRACT

Mfn2 is a mitochondrial fusion protein with bioenergetic functions implicated in the pathophysiology of neuronal and metabolic disorders. Understanding the bioenergetic mechanism of Mfn2 may aid in designing therapeutic approaches for these disorders. Here we show using endoplasmic reticulum (ER) or mitochondria-targeted Mfn2 that Mfn2 stimulation of the mitochondrial metabolism requires its localization in the ER, which is independent of its fusion function. ER-located Mfn2 interacts with mitochondrial Mfn1/2 to tether the ER and mitochondria together, allowing Ca2+ transfer from the ER to mitochondria to enhance mitochondrial bioenergetics. The physiological relevance of these findings is shown during neurite outgrowth, when there is an increase in Mfn2-dependent ER-mitochondria contact that is necessary for correct neuronal arbor growth. Reduced neuritic growth in Mfn2 KO neurons is recovered by the expression of ER-targeted Mfn2 or an artificial ER-mitochondria tether, indicating that manipulation of ER-mitochondria contacts could be used to treat pathologic conditions involving Mfn2.


Subject(s)
Endoplasmic Reticulum , GTP Phosphohydrolases , Endoplasmic Reticulum/metabolism , Energy Metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
7.
Dis Model Mech ; 14(1)2021 01 26.
Article in English | MEDLINE | ID: mdl-33735101

ABSTRACT

Congenital diaphragmatic hernia (CDH) is a relatively common developmental defect with considerable mortality and morbidity. Formation of the diaphragm is a complex process that involves several cell types, each with different developmental origins. Owing to this complexity, the aetiology of CDH is not well understood. The pleuroperitoneal folds (PPFs) and the posthepatic mesenchymal plate (PHMP) are transient structures that are essential during diaphragm development. Using several mouse models, including lineage tracing, we demonstrate the heterogeneous nature of the cells that make up the PPFs. The conditional deletion of Wilms tumor 1 homolog (Wt1) in the non-muscle mesenchyme of the PPFs results in CDH. We show that the fusion of the PPFs and the PHMP to form a continuous band of tissue involves movements of cells from both sources. The PPFs of mutant mice fail to fuse with the PHMP and exhibit increased RALDH2 (also known as ALDH1A2) expression. However, no changes in the expression of genes (including Snai1, Snai2, Cdh1 and Vim) implicated in epithelial-to-mesenchymal transition are observed. Additionally, the mutant PPFs lack migrating myoblasts and muscle connective tissue fibroblasts (TCF4+/GATA4+), suggesting possible interactions between these cell types. Our study demonstrates the importance of the non-muscle mesenchyme in development of the diaphragm.


Subject(s)
Diaphragm/pathology , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Animals , Connective Tissue , Disease Models, Animal , Female , Fibroblasts/metabolism , Gene Deletion , Gene Expression Profiling , Hernias, Diaphragmatic, Congenital/genetics , Male , Mice , Muscle Development , Time Factors , Transgenes , WT1 Proteins/metabolism
8.
Development ; 146(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31624071

ABSTRACT

The epicardium plays a crucial role in embryonic heart development and adult heart repair; however, the molecular events underlying its maturation remain unknown. Wt1, one of the main markers of the embryonic epicardium, is essential for epicardial development and function. Here, we analyse the transcriptomic profile of epicardial-enriched cells at different stages of development and from control and epicardial-specific Wt1 knockout (Wt1KO) mice. Transcriptomic and cell morphology analyses of epicardial cells from epicardial-specific Wt1KO mice revealed a defect in the maturation process of the mutant epicardium, including sustained upregulation of Bmp4 expression and the inability of mutant epicardial cells to transition into a mature squamous phenotype. We identified Bmp4 as a transcriptional target of Wt1, thus providing a molecular basis for the retention of the cuboidal cell shape observed in the Wt1KO epicardium. Accordingly, inhibition of the Bmp4 signalling pathway both ex vivo and in vivo rescued the cuboidal phenotype of the mutant epicardium. Our findings indicate the importance of the cuboidal-to-squamous transition in epicardial maturation, a process regulated by Wt1.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Pericardium/cytology , Pericardium/metabolism , WT1 Proteins/metabolism , Animals , Cell Shape/drug effects , Cell Shape/genetics , Cells, Cultured , Female , Flow Cytometry , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Heart/drug effects , Male , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Myocardium/metabolism , Myocardium/ultrastructure , Pericardium/drug effects , Pericardium/ultrastructure , WT1 Proteins/genetics
9.
EMBO J ; 37(9)2018 05 02.
Article in English | MEDLINE | ID: mdl-29615453

ABSTRACT

The formation of neurites is an important process affecting the cognitive abilities of an organism. Neurite growth requires the addition of new membranes, but the metabolic remodeling necessary to supply lipids for membrane expansion is poorly understood. Here, we show that synaptic activity, one of the most important inducers of neurite growth, transcriptionally regulates the expression of neuronal glucose transporter Glut3 and rate-limiting enzymes of glycolysis, resulting in enhanced glucose uptake and metabolism that is partly used for lipid synthesis. Mechanistically, CREB regulates the expression of Glut3 and Siah2, the latter and LDH activity promoting the normoxic stabilization of HIF-1α that regulates the expression of rate-limiting genes of glycolysis. The expression of dominant-negative HIF-1α or Glut3 knockdown blocks activity-dependent neurite growth in vitro while pharmacological inhibition of the glycolysis and specific ablation of HIF-1α in early postnatal mice impairs the neurite architecture. These results suggest that the manipulation of neuronal glucose metabolism could be used to treat some brain developmental disorders.


Subject(s)
Cell Membrane Structures/metabolism , Neurites/metabolism , Synapses/metabolism , Animals , Cell Membrane Structures/genetics , Cell Membrane Structures/pathology , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Glucose Transporter Type 3/biosynthesis , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Neurites/pathology , Rats , Rats, Sprague-Dawley , Synapses/genetics , Synapses/pathology , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
10.
Methods Mol Biol ; 1467: 61-71, 2016.
Article in English | MEDLINE | ID: mdl-27417959

ABSTRACT

The embryonic epicardium is an important source of cardiovascular precursor cells and paracrine factors required for adequate heart formation. During embryonic heart formation, WT1 is mainly expressed in epicardial cells and epicardial derived cells. Its expression has been used to trace epicardial derivatives in embryos and recently it has been used to follow the reactivation of epicardial cells after myocardial infarction. Interestingly, the highest level of expression of WT1 during epicardium development correlates with the highest proliferative state, stem cell properties, and migratory capacity of epicardial cells. Here, we review the various types of tools and strategies used to study WT1 function in the embryonic epicardium and provide examples of their use.


Subject(s)
Pericardium/cytology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Systems Biology/methods , Animals , Cell Movement , Cell Proliferation , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Expression Regulation, Developmental , Mice , Pericardium/embryology , Pericardium/metabolism , Stem Cells/cytology , Stem Cells/metabolism , WT1 Proteins
11.
Cell Cycle ; 14(9): 1365-9, 2015.
Article in English | MEDLINE | ID: mdl-25789413

ABSTRACT

Mitochondria morphology constantly changes through fission and fusion processes that regulate mitochondrial function, and it therefore plays a prominent role in cellular homeostasis. Cell death progression is associated with mitochondrial fission. Fission is mediated by the mainly cytoplasmic Drp1, which is activated by different post-translational modifications and recruited to mitochondria to perform its function. Our research and other studies have shown that in the early moments of excitotoxic insult Drp1 must be nitrosylated to mediate mitochondrial fragmentation in neurons. Nonetheless, mitochondrial fission is a multistep process in which filamentous actin assembly/disassembly and myosin-mediated mitochondrial constriction play prominent roles. Here we establish that in addition to nitric oxide production, excitotoxicity-induced mitochondrial fragmentation also requires activation of the actomyosin regulator ROCK. Although ROCK1 has been shown to phosphorylate and activate Drp1, experiments using phosphor-mutant forms of Drp1 in primary cortical neurons indicate that in excitotoxic conditions, ROCK does not act directly on Drp1 to mediate fission, but may act on the actomyosin complex. Thus, these data indicate that a wider range of signaling pathways than those that target Drp1 are amenable to be inhibited to prevent mitochondrial fragmentation as therapeutic option.


Subject(s)
Cerebral Cortex/drug effects , Excitatory Amino Acid Agonists/toxicity , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , N-Methylaspartate/toxicity , Neurons/drug effects , rho-Associated Kinases/metabolism , Actomyosin/metabolism , Animals , Cells, Cultured , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Dynamins/genetics , Dynamins/metabolism , Enzyme Activation , Mitochondria/enzymology , Mitochondria/pathology , Neurons/enzymology , Neurons/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transfection , rho-Associated Kinases/antagonists & inhibitors
12.
Nat Cell Biol ; 16(4): 367-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24609269

ABSTRACT

Fuelled by the obesity epidemic, there is considerable interest in the developmental origins of white adipose tissue (WAT) and the stem and progenitor cells from which it arises. Whereas increased visceral fat mass is associated with metabolic dysfunction, increased subcutaneous WAT is protective. There are six visceral fat depots: perirenal, gonadal, epicardial, retroperitoneal, omental and mesenteric, and it is a subject of much debate whether these have a common developmental origin and whether this differs from that for subcutaneous WAT. Here we show that all six visceral WAT depots receive a significant contribution from cells expressing Wt1 late in gestation. Conversely, no subcutaneous WAT or brown adipose tissue arises from Wt1-expressing cells. Postnatally, a subset of visceral WAT continues to arise from Wt1-expressing cells, consistent with the finding that Wt1 marks a proportion of cell populations enriched in WAT progenitors. We show that all visceral fat depots have a mesothelial layer like the visceral organs with which they are associated, and provide several lines of evidence that Wt1-expressing mesothelium can produce adipocytes. These results reveal a major ontogenetic difference between visceral and subcutaneous WAT, and pinpoint the lateral plate mesoderm as a major source of visceral WAT. They also support the notion that visceral WAT progenitors are heterogeneous, and suggest that mesothelium is a source of adipocytes.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , WT1 Proteins/metabolism , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/embryology , Adipose Tissue, White/cytology , Adipose Tissue, White/embryology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Lineage/genetics , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Tamoxifen/pharmacology , WT1 Proteins/genetics
13.
Hum Mol Genet ; 22(25): 5083-95, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-23900076

ABSTRACT

The embryonic epicardium is an important source of cardiovascular precursor cells and paracrine factors that are required for adequate heart formation. Signaling pathways regulated by WT1 that promote heart development have started to be described; however, there is little information on signaling pathways regulated by WT1 that could act in a negative manner. Transcriptome analysis of Wt1KO epicardial cells reveals an unexpected role for WT1 in repressing the expression of interferon-regulated genes that could be involved in a negative regulation of heart morphogenesis. Here, we showed that WT1 is required to repress the expression of the chemokines Ccl5 and Cxcl10 in epicardial cells. We observed an inverse correlation of Wt1 and the expression of Cxcl10 and Ccl5 during epicardium development. Chemokine receptor analyses of hearts from Wt1(gfp/+) mice demonstrate the differential expression of their chemokine receptors in GFP(+) epicardial enriched cells and GFP(-) cells. Functional assays demonstrate that CXCL10 and CCL5 inhibit epicardial cells migration and the proliferation of cardiomyocytes respectively. WT1 regulates the expression levels of Cxcl10 and Ccl5 in epicardial cells directly and indirectly through increasing the levels of IRF7. As epicardial cell reactivation after a myocardial damage is linked with WT1 expression, the present work has potential implications in adult heart repair.


Subject(s)
Chemokine CCL5/biosynthesis , Chemokine CXCL10/biosynthesis , Heart/growth & development , Pericardium/growth & development , WT1 Proteins/genetics , Animals , Chemokine CCL5/genetics , Chemokine CXCL10/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Interferon Regulatory Factor-7/metabolism , Mice , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pericardium/cytology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/metabolism , Signal Transduction , WT1 Proteins/biosynthesis
14.
Dev Cell ; 21(3): 559-74, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21871842

ABSTRACT

Wt1 regulates the epithelial-mesenchymal transition (EMT) in the epicardium and the reverse process (MET) in kidney mesenchyme. The mechanisms underlying these reciprocal functions are unknown. Here, we show in both embryos and cultured cells that Wt1 regulates Wnt4 expression dichotomously. In kidney cells, Wt1 recruits Cbp and p300 as coactivators; in epicardial cells it enlists Basp1 as a corepressor. Surprisingly, in both tissues, Wt1 loss reciprocally switches the chromatin architecture of the entire Ctcf-bounded Wnt4 locus, but not the flanking regions; we term this mode of action "chromatin flip-flop." Ctcf and cohesin are dispensable for Wt1-mediated chromatin flip-flop but essential for maintaining the insulating boundaries. This work demonstrates that a developmental regulator coordinates chromatin boundaries with the transcriptional competence of the flanked region. These findings also have implications for hierarchical transcriptional regulation in development and disease.

15.
Development ; 138(6): 1093-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21343363

ABSTRACT

Epicardial-derived signals are key regulators of cardiac embryonic development. An important part of these signals is known to relate to a retinoic acid (RA) receptor-dependent mechanism. RA is a potent morphogen synthesised by Raldh enzymes, Raldh2 being the predominant one in mesodermal tissues. Despite the importance of epicardial retinoid signalling in the heart, the molecular mechanisms controlling cardiac Raldh2 transcription remain unknown. In the current study, we show that Wt1-null epicardial cells display decreased expression of Raldh2 both in vivo and in vitro. Using a RA-responsive reporter, we have confirmed that Wt1-null epicardial cells actually show reduced synthesis of RA. We also demonstrate that Raldh2 is a direct transcriptional target of Wt1 in epicardial cells. A secondary objective of this study was to identify the status of RA-related receptors previously reported to be critical to epicardial biology (PDGFRα,ß; RXRα). PDGFRα and PDGFRß mRNA and protein levels are downregulated in the absence of Wt1, but only Pdgfra expression is rescued by the addition of RA to Wt1-null epicardial cells. RXRα mRNA levels are not affected in Wt1-null epicardial cells. Taken together, our results indicate that Wt1 critically regulates epicardial RA signalling via direct activation of the Raldh2 gene, and identify a role for Wt1 in the regulation of morphogen receptors involved in the proliferation, migration, and differentiation of epicardial and epicardially-derived cells (EPDC).


Subject(s)
Aldehyde Oxidoreductases/genetics , Pericardium/embryology , Tretinoin/metabolism , WT1 Proteins/physiology , Aldehyde Oxidoreductases/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Heart/embryology , Mice , Mice, Transgenic , Pericardium/metabolism , Signal Transduction/genetics , Transcriptional Activation/drug effects , Tretinoin/pharmacology , Tretinoin/physiology
16.
Nat Genet ; 42(1): 89-93, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20023660

ABSTRACT

The epicardial epithelial-mesenchymal transition (EMT) is hypothesized to generate cardiovascular progenitor cells that differentiate into various cell types, including coronary smooth muscle and endothelial cells, perivascular and cardiac interstitial fibroblasts and cardiomyocytes. Here we show that an epicardial-specific knockout of the gene encoding Wilms' tumor-1 (Wt1) leads to a reduction in mesenchymal progenitor cells and their derivatives. We show that Wt1 is essential for repression of the epithelial phenotype in epicardial cells and during embryonic stem cell differentiation through direct transcriptional regulation of the genes encoding Snail (Snai1) and E-cadherin (Cdh1), two of the major mediators of EMT. Some mesodermal lineages do not form in Wt1-null embryoid bodies, but this effect is rescued by the expression of Snai1, underscoring the importance of EMT in generating these differentiated cells. These new insights into the molecular mechanisms regulating cardiovascular progenitor cells and EMT will shed light on the pathogenesis of heart diseases and may help the development of cell-based therapies.


Subject(s)
Cadherins/genetics , Stem Cells/metabolism , Transcription Factors/genetics , Transcription, Genetic , WT1 Proteins/genetics , Animals , Cadherins/metabolism , Cardiovascular System/cytology , Cell Differentiation , Cells, Cultured , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epithelium/metabolism , Epithelium/pathology , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Knockout , Pericardium/abnormalities , Pericardium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Stem Cells/cytology , Transcription Factors/metabolism , WT1 Proteins/metabolism
17.
Blood Press ; 17(5-6): 298-305, 2008.
Article in English | MEDLINE | ID: mdl-19085535

ABSTRACT

BACKGROUND: Hypertension has been related to endothelial dysfunction. Patients with refractory hypertension (RH) have a reduced number of endothelial progenitor cells (EPCs). AIM: To evaluate if blood EPC levels relate to endothelium-dependent vasodilation (ED-VD) in RH. METHODS: We analyzed 29 RH confirmed by 24-h ambulatory blood pressure monitoring and assessed complete clinical and laboratory evaluation. EPCs were isolated from peripheral mononuclear cells (MNC) by flow cytometry. ED-VD was determined measuring flow-mediated dilation (FMD) by venous occlusion plethysmography. Results. Circulating EPCs/10(5) MNC (median [Q1-Q3]): 23.0 [4.5-53.8]. FMD (median [Q1-Q3]): 211.7 [79.5-365.8]%. Significant correlations with log-FMD: EPCs (r = 0.469; p = 0.018) and homocysteine (r = -0.414; p = 0.045). There was no collinearity between EPCs and homocysteine. FMD did not correlate with age, gender, office BP, 24-h systolic blood pressure or 24-h diastolic blood pressure, laboratory parameters, C-reactive-protein, left ventricular-mass index, dyslipidaemia, smoking habit and statin or angiotensin system blockers treatment. Multiple linear regression analysis showed that after age-adjustment, EPC (p = 0.027) and homocysteine (p = 0.004) were the only variables that predicted FMD (R = 0.740). After dividing patients according to EPC number, patients in the lower tertile showed a significantly reduced FMD compared with those in the group of the two upper tertiles of EPC: log-FMD (mean+/-SD): 4.7+/-0.9 vs 5.6+/-0.8, respectively (p = 0.031). CONCLUSIONS: ED-VD independently correlates with circulating EPCs in RH. Homocysteine is also an independent predictor of lower FMD in such patients.


Subject(s)
Dilatation, Pathologic/diagnosis , Endothelial Cells/pathology , Hypertension/pathology , Stem Cells/pathology , Adult , Cell Count , Cell Separation , Dilatation, Pathologic/pathology , Flow Cytometry , Homocysteine/blood , Humans , Middle Aged , Predictive Value of Tests , Regression Analysis , Vasodilation
18.
Biochem J ; 394(Pt 2): 449-57, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16232121

ABSTRACT

Claudin-1 is an integral membrane protein component of tight junctions. The Snail family of transcription factors are repressors that play a central role in the epithelial-mesenchymal transition, a process that occurs during cancer progression. Snail and Slug members are direct repressors of E-cadherin and act by binding to the specific E-boxes of its proximal promoter. In the present study, we demonstrate that overexpression of Slug or Snail causes a decrease in transepithelial electrical resistance. Overexpression of Slug and Snail in MDCK (Madin-Darby canine kidney) cells down-regulated Claudin-1 at protein and mRNA levels. In addition, Snail and Slug are able to effectively repress human Claudin-1-driven reporter gene constructs containing the wild-type promoter sequence, but not those with mutations in two proximal E-box elements. We also demonstrate by band-shift assay that Snail and Slug bind to the E-box motifs present in the human Claudin-1 promoter. Moreover, an inverse correlation in the levels of Claudin-1 and Slug transcripts were observed in breast cancer cell lines. E-box elements in the Claudin-1 promoter were found to play a critical negative regulatory role in breast cancer cell lines that expressed low levels of Claudin-1 transcript. Significantly, in invasive human breast tumours, high levels of Snail and Slug correlated with low levels of Claudin-1 expression. Taken together, these results support the hypothesis that Claudin-1 is a direct downstream target gene of Snail family factors in epithelial cells.


Subject(s)
Down-Regulation , Epithelial Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line , Claudin-1 , Dogs , Fibroblasts/metabolism , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Snail Family Transcription Factors
19.
Am J Physiol Lung Cell Mol Physiol ; 288(6): L1081-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15886398

ABSTRACT

Junctional adhesion molecule-A (JAM-A) regulates key inflammatory responses, such as edema formation and leukocyte transmigration. Although it has been reported that the inflammatory cytokine tumor necrosis factor (TNF) causes the disassembly of JAM-A from the intercellular junctions, the mechanism has not been elucidated fully. Here, we report that TNF enhances the solubility of JAM-A in Triton X-100 and increases the amount of Triton-soluble JAM-A dimers at the cell surface but does not change the total levels of cellular JAM-A. Thus we hypothesized that TNF causes the redistribution of JAM-A from the junctions to the cell surface and that junction disassembly is sufficient to account for JAM-A redistribution. Intriguingly, however, even after complete disassembly of the junctions (with EDTA and trypsin), higher levels of JAM-A are detectable at the cell surface (by FACS analysis) in cells that had been previously incubated in the presence of TNF than in its absence. Thus we propose that TNF causes not only the disassembly of JAM-A from the junctions and its subsequent redistribution to the cell surface but also its dispersal in such a way that JAM-A becomes more easily accessible to the antibodies used for FACS analysis. Finally, we evaluated whether soluble fibronectin might attenuate the effects of TNF on JAM-A, as some inflammatory conditions are associated with the depletion of plasma fibronectin. We found that fibronectin reduces the effect of TNF on the disassembly of JAM-A, but not on its dispersal, thus further stressing that disassembly and dispersal can be functionally dissociated.


Subject(s)
Cell Adhesion Molecules/metabolism , Endothelium, Vascular/drug effects , Fibronectins/pharmacology , Intercellular Junctions/drug effects , Tight Junctions/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Intercellular Junctions/metabolism , Junctional Adhesion Molecules , Mice , Octoxynol , Solubility , Tight Junctions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...