Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Matrix Biol ; 87: 11-25, 2020 05.
Article in English | MEDLINE | ID: mdl-31422156

ABSTRACT

Multimerin-2 is an extracellular matrix glycoprotein and member of the elastin microfibril interface-located (EMILIN) family of proteins. Multimerin-2 is deposited along blood vessels and we previously demonstrated that it regulates the VEGFA/VEGFR2 signaling axis and angiogenesis. However, its role in modulating vascular homeostasis remains largely unexplored. Here we identified Multimerin-2 as a key molecule required to maintain vascular stability. RNAi knockdown of Multimerin-2 in endothelial cells led to cell-cell junctional instability and increased permeability. Mechanistically cell-cell junction dismantlement occurred through the phosphorylation of VEGFR2 at Tyr951, activation of Src and phosphorylation of VE-cadherin. To provide an in vivo validation for these in vitro effects, we generated Multimerin-2-/- (Mmrn2-/-) mice. Although Mmrn2-/- mice developed normally and displayed no gross abnormalities, endothelial cells displayed cell junctional defects associated with increased levels of VEGFR2 phospho-Tyr949 (the murine counterpart of human Tyr951), impaired pericyte recruitment and increased vascular leakage. Of note, tumor associated vessels were defective in Mmrn2-/- mice, with increased number of small and often collapsed vessels, concurrent with a significant depletion of pericytic coverage. Consequently, the Mmrn2-/- vessels were less perfused and leakier, leading to increased tumor hypoxia. Chemotherapy efficacy was markedly impaired in Mmrn2-/- mice and this was associated with poor drug delivery to the tumor xenografts. Collectively, our findings demonstrate that Multimerin-2 is required for proper vessel homeostasis and stabilization, and unveil the possibility to utilize expression levels of this glycoprotein in predicting chemotherapy efficacy.


Subject(s)
Antigens, CD/metabolism , Antigens, Surface/genetics , Cadherins/metabolism , Extracellular Matrix Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Melanoma/blood supply , Membrane Glycoproteins/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/pharmacology , Drug Therapy , Extracellular Matrix Proteins/metabolism , Gene Knockout Techniques , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions , Intercellular Signaling Peptides and Proteins/metabolism , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Mice , Neoplasm Transplantation , Phosphorylation , Tumor Hypoxia/drug effects
2.
Arterioscler Thromb Vasc Biol ; 38(10): 2484-2497, 2018 10.
Article in English | MEDLINE | ID: mdl-30354220

ABSTRACT

Objective- EMILIN-1 (elastin microfibrils interface located protein-1) protein inhibits pro-TGF-ß (transforming growth factor-ß) proteolysis and limits TGF-ß bioavailability in vascular extracellular matrix. Emilin1-/- null mice display increased vascular TGF-ß signaling and are hypertensive. Because EMILIN-1 is expressed in vessels from embryonic life to adulthood, we aimed at unravelling whether the hypertensive phenotype of Emilin1-/- null mice results from a developmental defect or lack of homeostatic role in the adult. Approach and Results- By using a conditional gene targeting inactivating EMILIN-1 in smooth muscle cells of adult mice, we show that increased blood pressure in mice with selective smooth muscle cell ablation of EMILIN-1 depends on enhanced myogenic tone. Mechanistically, we unveil that higher TGF-ß signaling in smooth muscle cells stimulates HB-EGF (heparin-binding epidermal growth factor) expression and subsequent transactivation of EGFR (epidermal growth factor receptor). With increasing intraluminal pressure in resistance arteries, the cross talk established by TGF-ß and EGFR signals recruits TRPC6 (TRP [transient receptor potential] classical type 6) and TRPM4 (TRP melastatin type 4) channels, lastly stimulating voltage-dependent calcium channels and potentiating myogenic tone. We found reduced EMILIN-1 and enhanced myogenic tone, dependent on increased TGF-ß-EGFR signaling, in resistance arteries from hypertensive patients. Conclusions- Taken together, our findings implicate an unexpected role of the TGF-ß-EGFR pathway in hypertension with current translational perspectives.


Subject(s)
ErbB Receptors/metabolism , Hypertension/metabolism , Membrane Glycoproteins/metabolism , Mesenteric Arteries/metabolism , Transforming Growth Factor beta1/metabolism , Vasoconstriction , Animals , Blood Pressure , Calcium Channels/metabolism , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Female , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Hypertension/genetics , Hypertension/physiopathology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Signal Transduction , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , TRPM Cation Channels/metabolism , Transforming Growth Factor beta1/pharmacology , Vasoconstriction/drug effects
3.
Oncogene ; 37(25): 3399-3414, 2018 06.
Article in English | MEDLINE | ID: mdl-29483644

ABSTRACT

EMILIN2 is an extracellular matrix constituent playing an important role in angiogenesis; however, the underlying mechanism is unknown. Here we show that EMILIN2 promotes angiogenesis by directly binding epidermal growth factor receptor (EGFR), which enhances interleukin-8 (IL-8) production. In turn, IL-8 stimulates the proliferation and migration of vascular endothelial cells. Emilin2 null mice were generated and exhibited delayed retinal vascular development, which was rescued by the administration of the IL-8 murine ortholog MIP-2. Next, we assessed tumor growth and tumor-associated angiogenesis in these mice. Tumor cell growth in Emilin2 null mice was impaired as well as the expression of MIP-2. The vascular density of the tumors developed in Emilin2 null mice was prejudiced and vessels perfusion, as well as response to chemotherapy, decreased. Accordingly, human tumors expressing high levels of EMILIN2 were more responsive to chemotherapy. These results point at EMILIN2 as a key microenvironmental cue affecting vessel formation and unveil the possibility to develop new prognostic tools to predict chemotherapy efficacy.


Subject(s)
Glycoproteins/metabolism , Glycoproteins/physiology , Interleukin-8/metabolism , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Neovascularization, Pathologic/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Glycoproteins/genetics , Humans , Interleukin-8/genetics , Male , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Rats , Rats, Inbred F344 , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Immunity ; 47(5): 959-973.e9, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29150241

ABSTRACT

Aortic aneurysms are life-threatening conditions with effective treatments mainly limited to emergency surgery or trans-arterial endovascular stent grafts, thus calling for the identification of specific molecular targets. Genetic studies have highlighted controversial roles of transforming growth factor ß (TGF-ß) signaling in aneurysm development. Here, we report on aneurysms developing in adult mice after smooth muscle cell (SMC)-specific inactivation of Smad4, an intracellular transducer of TGF-ß. The results revealed that Smad4 inhibition activated interleukin-1ß (IL-1ß) in SMCs. This danger signal later recruited innate immunity in the adventitia through chemokine (C-C motif) ligand 2 (CCL2) and modified the mechanical properties of the aortic wall, thus favoring vessel dilation. SMC-specific Smad4 deletion in Il1r1- or Ccr2-null mice resulted in milder aortic pathology. A chronic treatment with anti-IL-1ß antibody effectively hampered aneurysm development. These findings identify a mechanistic target for controlling the progression of aneurysms with compromised TGF-ß signaling, such as those driven by SMAD4 mutations.


Subject(s)
Aortic Aneurysm/prevention & control , Interleukin-1beta/antagonists & inhibitors , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Cells, Cultured , Chemokine CCL2/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Mice , Myocytes, Smooth Muscle/immunology , NF-kappa B/physiology , Receptors, CCR2/antagonists & inhibitors , Smad4 Protein/physiology , Tamoxifen/pharmacology
5.
Physiol Rep ; 5(5)2017 Mar.
Article in English | MEDLINE | ID: mdl-28270590

ABSTRACT

Aortic valve disease (AVD) is a common condition with a progressive natural history, and presently, there are no pharmacologic treatment strategies. Elastic fiber fragmentation (EFF) is a hallmark of AVD, and increasing evidence implicates developmental elastic fiber assembly defects. Emilin1 is a glycoprotein necessary for elastic fiber assembly that is present in both developing and mature human and mouse aortic valves. The Emilin1-deficient mouse (Emilin1-/- ) is a model of latent AVD, characterized by activated TGFß/MEK/p-Erk signaling and upregulated elastase activity. Emilin1-/- aortic valves demonstrate early EFF and aberrant angiogenesis followed by late neovascularization and fibrosis. The objective of this study was to test the effectiveness of three different targeted therapies. Aged (12-14 months) Emilin1-/- mice were treated with refametinib (RDEA-119, MEK1/2 inhibitor), doxycycline (elastase inhibitor), or G6-31 (anti-VEGF-A mouse antibody) for 4 weeks. Refametinib- and doxycycline-treated Emilin1-/- mice markedly reduced MEK/p-Erk activation in valve tissue. Furthermore, both refametinib and doxycycline attenuated elastolytic cathepsin K, L, MMP-2, and MMP-9 activation, and abrogated macrophage and neutrophil infiltration in Emilin1-/- aortic valves. RNAseq analysis was performed in aortic valve tissue from adult (4 months) and aged (14 months) Emilin1-/- and age-matched wild-type control mice, and demonstrated upregulation of genes associated with MAPK/MEK/p-Erk signaling and elastases at the adult stage and inflammatory pathways at the aged stage controlling for age. These results suggest that Erk1/2 signaling is an important modulator of early elastase activation, and pharmacological inhibition using refametinib may be a promising treatment to halt AVD progression.


Subject(s)
Antibodies/therapeutic use , Aortic Valve/drug effects , Diphenylamine/analogs & derivatives , Doxycycline/therapeutic use , Heart Defects, Congenital/drug therapy , Heart Valve Diseases/drug therapy , MAP Kinase Signaling System/drug effects , Membrane Glycoproteins/genetics , Sulfonamides/therapeutic use , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies/pharmacology , Aortic Valve/metabolism , Aortic Valve/pathology , Bicuspid Aortic Valve Disease , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Disease Models, Animal , Disease Progression , Doxycycline/pharmacology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Mice , Mice, Knockout , Pancreatic Elastase/metabolism , Sulfonamides/pharmacology , Up-Regulation/drug effects
6.
Dis Model Mech ; 7(8): 987-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25056700

ABSTRACT

Aortic valve disease (AVD) is characterized by elastic fiber fragmentation (EFF), fibrosis and aberrant angiogenesis. Emilin1 is an elastin-binding glycoprotein that regulates elastogenesis and inhibits TGF-ß signaling, but the role of Emilin1 in valve tissue is unknown. We tested the hypothesis that Emilin1 deficiency results in AVD, mediated by non-canonical (MAPK/phosphorylated Erk1 and Erk2) TGF-ß dysregulation. Using histology, immunohistochemistry, electron microscopy, quantitative gene expression analysis, immunoblotting and echocardiography, we examined the effects of Emilin1 deficiency (Emilin1-/-) in mouse aortic valve tissue. Emilin1 deficiency results in early postnatal cell-matrix defects in aortic valve tissue, including EFF, that progress to latent AVD and premature death. The Emilin1-/- aortic valve displays early aberrant provisional angiogenesis and late neovascularization. In addition, Emilin1-/- aortic valves are characterized by early valve interstitial cell activation and proliferation and late myofibroblast-like cell activation and fibrosis. Interestingly, canonical TGF-ß signaling (phosphorylated Smad2 and Smad3) is upregulated constitutively from birth to senescence, whereas non-canonical TGF-ß signaling (phosphorylated Erk1 and Erk2) progressively increases over time. Emilin1 deficiency recapitulates human fibrotic AVD, and advanced disease is mediated by non-canonical (MAPK/phosphorylated Erk1 and Erk2) TGF-ß activation. The early manifestation of EFF and aberrant angiogenesis suggests that these processes are crucial intermediate factors involved in disease progression and therefore might provide new therapeutic targets for human AVD.


Subject(s)
Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Membrane Glycoproteins/deficiency , Neovascularization, Pathologic/metabolism , Transforming Growth Factor beta/metabolism , Animals , Aortic Valve/diagnostic imaging , Aortic Valve/metabolism , Aortic Valve/pathology , Aortic Valve/ultrastructure , Bicuspid Aortic Valve Disease , Calcinosis/complications , Calcinosis/pathology , Cell Proliferation , Cutis Laxa/pathology , Disease Models, Animal , Disease Progression , Elastic Tissue/metabolism , Fibrosis , Heart Defects, Congenital/complications , Heart Defects, Congenital/diagnostic imaging , Heart Valve Diseases/complications , Heart Valve Diseases/diagnostic imaging , Inflammation/complications , Inflammation/pathology , Membrane Glycoproteins/metabolism , Mice , Models, Biological , Myofibroblasts/metabolism , Myofibroblasts/pathology , Neovascularization, Pathologic/pathology , Signal Transduction , Ultrasonography
7.
Arterioscler Thromb Vasc Biol ; 32(9): 2178-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22814752

ABSTRACT

OBJECTIVE: Emilin-1 is a protein of elastic extracellular matrix involved in blood pressure (BP) control by negatively affecting transforming growth factor (TGF)-ß processing. Emilin1 null mice are hypertensive. This study investigates how Emilin-1 deals with vascular mechanisms regulating BP. METHODS AND RESULTS: This study uses a phenotype rescue approach in which Emilin-1 is expressed in either endothelial cells or vascular smooth muscle cells of transgenic animals with the Emilin1(-/-) background. We found that normalization of BP required Emilin-1 expression in smooth muscle cells, whereas expression of the protein in endothelial cells did not modify the hypertensive phenotype of Emilin1(-/-) mice. We also explored the effect of treatment with anti-TGF-ß antibodies on the hypertensive phenotype of Emilin1(-/-) mice, finding that neutralization of TGF-ß in Emilin1 null mice normalized BP quite rapidly (2 weeks). Finally, we evaluated the vasoconstriction response of resistance arteries to perfusion pressure and neurohumoral agents in different transgenic mouse lines. Interestingly, we found that the hypertensive phenotype was coupled with an increased arteriolar myogenic response to perfusion pressure, while the vasoconstriction induced by neurohumoral agents remained unaffected. We further elucidate that, as for the hypertensive phenotype, the increased myogenic response was attributable to increased TGF-ß activity. CONCLUSIONS: Our findings clarify that Emilin-1 produced by vascular smooth muscle cells acts as a main regulator of resting BP levels by controlling the myogenic response in resistance arteries through TGF-ß.


Subject(s)
Blood Pressure , Hypertension/metabolism , Membrane Glycoproteins/metabolism , Muscle, Smooth, Vascular/metabolism , Vasoconstriction , Animals , Antibodies, Neutralizing/administration & dosage , Arterioles/metabolism , Arterioles/physiopathology , Blood Pressure/drug effects , Blood Pressure/genetics , Blood Pressure Monitoring, Ambulatory/methods , Dose-Response Relationship, Drug , Echocardiography, Doppler , Endothelial Cells/metabolism , Gene Expression Regulation , Genotype , Humans , Hypertension/genetics , Hypertension/physiopathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phenotype , Telemetry , Time Factors , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Vasoconstriction/drug effects , Vasoconstriction/genetics , Vasoconstrictor Agents/pharmacology
8.
J Biol Chem ; 287(14): 11498-515, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334695

ABSTRACT

EMILIN-3 is a glycoprotein of the extracellular matrix belonging to a family that contains a characteristic N-terminal cysteine-rich EMI domain. Currently, EMILIN-3 is the least characterized member of the elastin microfibril interface-located protein (EMILIN)/Multimerin family. Using RNA, immunohistochemical, and protein chemistry approaches, we carried out a detailed characterization of the expression and biochemical properties of EMILIN-3 in mouse. During embryonic and postnatal development, EMILIN-3 showed a peculiar and dynamic pattern of gene expression and protein distribution. EMILIN-3 mRNA was first detected at E8.5-E9.5 in the tail bud and in the primitive gut, and at later stages it became abundant in the developing gonads and osteogenic mesenchyme. Interestingly and in contrast to other EMILIN/Multimerin genes, EMILIN-3 was not found in the cardiovascular system. Despite the absence of the globular C1q domain, immunoprecipitation and Western blot analyses demonstrated that EMILIN-3 forms disulfide-bonded homotrimers and higher order oligomers. Circular dichroism spectroscopy indicated that the most C-terminal part of EMILIN-3 has a substantial α-helical content and forms coiled coil structures involved in EMILIN-3 homo-oligomerization. Transfection experiments with recombinant constructs showed that the EMI domain contributes to the higher order self-assembly but was dispensable for homotrimer formation. EMILIN-3 was found to bind heparin with high affinity, a property mediated by the EMI domain, thus revealing a new function for this domain that may contribute to the interaction of EMILIN-3 with other extracellular matrix and/or cell surface molecules. Finally, in vitro experiments showed that EMILIN-3 is able to function as an extracellular regulator of the activity of TGF-ß ligands.


Subject(s)
Antigens, Surface/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Developmental , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Protein Multimerization , Transforming Growth Factor beta/antagonists & inhibitors , Amino Acid Sequence , Animals , Antigens, Surface/chemistry , Antigens, Surface/genetics , Disulfides/chemistry , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Glycoproteins/chemistry , Glycoproteins/genetics , HEK293 Cells , Heparin/metabolism , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Molecular Sequence Data , Molecular Weight , Polysaccharides/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Transport
9.
Front Immunol ; 2: 93, 2011.
Article in English | MEDLINE | ID: mdl-22566882

ABSTRACT

Elastin microfibrillar interface proteins (EMILINs) and Multimerins (EMILIN1, EMILIN2, Multimerin1, and Multimerin2) constitute a four member family that in addition to the shared C-terminus gC1q domain typical of the gC1q/TNF superfamily members contain a N-terminus unique cysteine-rich EMI domain. These glycoproteins are homotrimeric and assemble into high molecular weight multimers. They are predominantly expressed in the extracellular matrix and contribute to several cellular functions in part associated with the gC1q domain and in part not yet assigned nor linked to other specific regions of the sequence. Among the latter is the control of arterial blood pressure, the inhibition of Bacillus anthracis cell cytotoxicity, the promotion of cell death, the proangiogenic function, and a role in platelet hemostasis. The focus of this review is to highlight the multiplicity of functions and domains of the EMILIN/Multimerin family with a particular emphasis on the regulatory role played by the ligand-receptor interactions of the gC1q domain. EMILIN1 is the most extensively studied member both from the structural and functional point of view. The structure of the gC1q of EMILIN1 solved by NMR highlights unique characteristics compared to other gC1q domains: it shows a marked decrease of the contact surface of the trimeric assembly and while conserving the jelly-roll topology with two ß-sheets of antiparallel strands it presents a nine-stranded ß-sandwich fold instead of the usual 10-stranded fold. This is likely due to the insertion of nine residues that disrupt the ordered strand organization and forma a highly dynamic protruding loop. In this loop the residue E933 is the site of interaction between gC1q and the α4ß1 and α9ß1 integrins, and contrary to integrin occupancy that usually upregulates cell growth, when gC1q is ligated by the integrin the cells reduce their proliferative activity.

10.
Development ; 137(15): 2571-8, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20573697

ABSTRACT

The definition of embryonic potency and induction of specific cell fates are intimately linked to the tight control over TGFbeta signaling. Although extracellular regulation of ligand availability has received considerable attention in recent years, surprisingly little is known about the intracellular factors that negatively control Smad activity in mammalian tissues. By means of genetic ablation, we show that the Smad4 inhibitor ectodermin (Ecto, also known as Trim33 or Tif1gamma) is required to limit Nodal responsiveness in vivo. New phenotypes, which are linked to excessive Nodal activity, emerge from such a modified landscape of Smad responsiveness in both embryonic and extra-embryonic territories. In extra-embryonic endoderm, Ecto is required to confine expression of Nodal antagonists to the anterior visceral endoderm. In trophoblast cells, Ecto precisely doses Nodal activity, balancing stem cell self-renewal and differentiation. Epiblast-specific Ecto deficiency shifts mesoderm fates towards node/organizer fates, revealing the requirement of Smad inhibition for the precise allocation of cells along the primitive streak. This study unveils that intracellular negative control of Smad function by ectodermin/Tif1gamma is a crucial element in the cellular response to TGFbeta signals in mammalian tissues.


Subject(s)
Gene Expression Regulation, Developmental , Smad Proteins/metabolism , Transcription Factors/metabolism , Alleles , Animals , Body Patterning , Cell Differentiation , Crosses, Genetic , Ectoderm/metabolism , Mesoderm/metabolism , Mice , Models, Biological , Phenotype , Signal Transduction , Transforming Growth Factor beta/metabolism
11.
J Biotechnol ; 141(1-2): 8-17, 2009 Apr 20.
Article in English | MEDLINE | ID: mdl-19428725

ABSTRACT

RNA interference (RNAi) through the use of lentiviral vectors is a valuable technique to induce loss of function mutations in mammals. Although very promising, the method has found only limited application and its general applicability remains to be established. Here we analyze how different factors influence RNAi mediated silencing of Col6a1, a gene of the extracellular matrix with a complex pattern of tissue specific expression. Our results, obtained with vectors pLVTHM and pLVPT-rtTRKRAB, point out three parameters as major determinants of the efficiency of interference: the choice of interfering sequence, the number of proviral copies integrated into the mouse genome and the site of insertion of the provirus. Although low copy number may produce efficient interference with low frequency, the general trend is that the number of integrated proviral copies determines the level of silencing and the severity of phenotypic traits. The site of insertion not only determines the overall intensity of expression of the small interfering RNA (siRNA), but also introduces slight variability of silencing in different organs. A lentiviral vector (pLVPT-rtTRKRAB) with doxycycline-inducible production of siRNA was also tested. Control of expression by the drug was stringent in many tissues; however, in some tissues turning off of siRNA synthesis was not complete. The data support the application of lentiviral vectors used here in transgenesis.


Subject(s)
Collagen Type VI/metabolism , Lentivirus/genetics , RNA Interference , Animals , Apoptosis , Blotting, Northern , Cell Line , Collagen Type VI/genetics , Female , Gene Expression Profiling , Genetic Vectors/genetics , Immunoblotting , Male , Membrane Potential, Mitochondrial , Mice , Mice, Transgenic , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction
12.
Exp Cell Res ; 314(19): 3508-18, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18761340

ABSTRACT

Collagen VI is a survival factor for skeletal muscle produced by endomysial cells and localized in connective tissue around muscle fibers. Mutations of its genes (COL6A1, COL6A2 and COL6A3) cause two muscular disorders, Bethlem myopathy and Ullrich disease. Expression of Collagen VI is highly dynamic during development, suggesting that developmental and homeostatic cues of the muscle microenvironment are relevant to confine its expression in this tissue. In face of the large body of work highlighting the relevance for human diseases of the adhesion of muscle cells with their surrounding extracellular matrix, remarkably little is known on how myogenic cells control gene expression in the connective tissue cells that produce such matrix. By expressing promoter-lacZ constructs in transgenic mice, we identify a Col6a1 gene enhancer region that is necessary for activation of transcription in connective tissue cells associated with skeletal muscle. By means of a lacZ transgenic mouse line crossed in metD/D mutant background, in which muscles of limb buds fail to form, we provide evidence that the presence of cells of the myogenic lineage is needed for enhancer activation in mesenchymal cells. Accordingly, lack of myogenic cells in limb buds of metD/D mice reduces Collagen VI deposition in connective tissue. The Col6a1 enhancer characterized here is conserved in mammals and may be relevant in some cases of heritable diseases of Collagen VI.


Subject(s)
Collagen Type VI/genetics , Connective Tissue Cells/metabolism , Enhancer Elements, Genetic/genetics , Muscle Cells/metabolism , Transcriptional Activation , Animals , Collagen Type VI/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Signal Transduction
13.
Mol Cell Biol ; 28(12): 4026-39, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18411305

ABSTRACT

Lymphatic-vasculature function critically depends on extracellular matrix (ECM) and on its connections with lymphatic endothelial cells (LECs). However, the composition and the architecture of ECM have not been fully taken into consideration in studying the biology and the pathology of the lymphatic system. EMILIN1, an elastic microfibril-associated protein, is highly expressed by LECs in vitro and colocalizes with lymphatic vessels in several mouse tissues. A comparative study between WT and Emilin1-/- mice highlighted the fact that Emilin1 deficiency in both CD1 and C57BL/6 backgrounds results in hyperplasia, enlargement, and frequently an irregular pattern of superficial and visceral lymphatic vessels and in a significant reduction of anchoring filaments. Emilin1-deficient mice also develop larger lymphangiomas than WT mice. Lymphatic vascular morphological alterations are accompanied by functional defects, such as mild lymphedema, a highly significant drop in lymph drainage, and enhanced lymph leakage. Our findings demonstrate that EMILIN1 is involved in the regulation of the growth and in the maintenance of the integrity of lymphatic vessels, a fundamental requirement for efficient function. The phenotype displayed by Emilin1(-/-) mice is the first abnormal lymphatic phenotype associated with the deficiency of an ECM protein and identifies EMILIN1 as a novel local regulator of lymphangiogenesis.


Subject(s)
Gene Expression Regulation , Lymphangiogenesis , Lymphatic Vessels/metabolism , Membrane Glycoproteins/physiology , Animals , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Lymphatic Vessels/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phenotype
14.
Cell ; 124(5): 929-42, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16530041

ABSTRACT

TGF-beta proteins are main regulators of blood vessel development and maintenance. Here, we report an unprecedented link between TGF-beta signaling and arterial hypertension based on the analysis of mice mutant for Emilin1, a cysteine-rich secreted glycoprotein expressed in the vascular tree. Emilin1 knockout animals display increased blood pressure, increased peripheral vascular resistance, and reduced vessel size. Mechanistically, we found that Emilin1 inhibits TGF-beta signaling by binding specifically to the proTGF-beta precursor and preventing its maturation by furin convertases in the extracellular space. In support of these findings, genetic inactivation of Emilin1 causes increased TGF-beta signaling in the vascular wall. Strikingly, high blood pressure observed in Emilin1 mutants is rescued to normal levels upon inactivation of a single TGF-beta1 allele. This study highlights the importance of modulation of TGF-beta availability in the pathogenesis of hypertension.


Subject(s)
Blood Pressure/physiology , Homeostasis , Membrane Glycoproteins/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Arteries/cytology , Arteries/metabolism , Furin/metabolism , Gene Dosage , Genes, Reporter , Humans , Hypertension/etiology , Hypertension/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nodal Protein , Phenotype , Protein Precursors/metabolism , Protein Structure, Tertiary , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism
15.
J Biol Chem ; 280(16): 15749-60, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15705587

ABSTRACT

The location of regions that regulate transcription of the murine Emilin1 gene was investigated in a DNA fragment of 16.8 kb, including the entire gene and about 8.7 and 0.6 kb of 5'- and 3'-flanking sequences, respectively. The 8.7-kb segment contains the 5'-end of the putative 2310015E02Rik gene and the sequence that separates it from Emilin1, whereas the 0.6-kb fragment covers the region between Emilin1 and Ketohexokinase genes. Sequence comparison between species identified several conserved regions in the 5'-flanking sequence. Most of them contained chromatin DNase I-hypersensitive sites, which were located at about -950 (HS1), -3100 (HS2), -4750 (HS3), and -5150 (HS4) in cells expressing Emilin1 mRNA. Emilin1 transcription initiates at multiple sites, the major of which correspond to two Initiator sequences. Promoter assays suggest that core promoter activity was mainly dependent on Initiator1 and on Sp1-binding sites close to the Initiators. Moreover, one important regulatory region was contained between -1 and -169 bp and a second one between -630 bp and -1.1 kb. The latter harbors a putative binding site for transcription factor AP1 matching the location of HS1. The function of different regions was studied by expressing lacZ constructs in transgenic mice. The results show that the 16.8-kb segment contains regulatory sequences driving high level transcription in all the tissues where Emilin1 is expressed. Moreover, the data suggest that transcription in different tissues is achieved through combinatorial cooperation between various regions, rather than being dependent on a single cis-activating region specific for each tissue.


Subject(s)
Gene Expression Regulation , Genes, Regulator , Membrane Glycoproteins/genetics , Transcription, Genetic , 3' Flanking Region , 5' Flanking Region , Animals , Base Sequence , Genes, Reporter , Mice , Molecular Sequence Data , Organ Specificity , Promoter Regions, Genetic
16.
Matrix Biol ; 22(7): 549-56, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14996434

ABSTRACT

The EDEN gene superfamily comprises genes that contain the EMI domain, a structural motif recently identified in proteins of the extracellular matrix. We report here the detailed expression pattern of genes of the EMILIN/Multimerin family, the most numerous group of EDEN superfamily, during mouse development. In situ hybridization has revealed that the EMILIN/Multimerin genes are particularly expressed in the cardio-vascular system and in mesenchymal cells. In general, the territories of expression of each gene are partially overlapping or complementary with that of other members of the family and, usually, more than one gene of the family is active in different tissues, consistent with the possibility of functional compensation. The analysis is particularly relevant in the interpretation of gene targeting experiments.


Subject(s)
Animals, Newborn/metabolism , Blood Proteins/metabolism , Embryo, Mammalian/metabolism , Membrane Glycoproteins/metabolism , Aging/physiology , Animals , Animals, Newborn/growth & development , Blood Proteins/genetics , Cardiovascular System/embryology , Cardiovascular System/metabolism , Embryonic and Fetal Development , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Membrane Glycoproteins/genetics , Mesoderm/cytology , Mesoderm/metabolism , Mice , Multigene Family , Protein Structure, Tertiary , Terminology as Topic , Tissue Distribution
17.
Mol Cell Biol ; 24(2): 638-50, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14701737

ABSTRACT

EMILINs constitute a family of genes of the extracellular matrix with high structural similarity. Four genes have been identified so far in human and mouse. To gain insight into the function of this gene family, EMILIN-1 has been inactivated in the mouse by gene targeting. The homozygous animals were fertile and did not show obvious abnormalities. However, histological and ultrastructural examination revealed alterations of elastic fibers in aorta and skin. Formation of elastic fibers by mutant embryonic fibroblasts in culture was also abnormal. Additional alterations were observed in cell morphology and anchorage of endothelial and smooth muscle cells to elastic lamellae. Considering that EMILIN-1 is adhesive for cells and that the protein binds to elastin and fibulin-5, EMILIN-1 may regulate elastogenesis and vascular cell maintenance by stabilizing molecular interactions between elastic fiber components and by endowing elastic fibers with specific cell adhesion properties.


Subject(s)
Blood Vessels/abnormalities , Elastic Tissue/abnormalities , Extracellular Matrix Proteins/deficiency , Membrane Glycoproteins/deficiency , Animals , Blood Vessels/pathology , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cells, Cultured , Elastic Tissue/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/physiology , Female , Humans , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron
18.
Biochem Biophys Res Commun ; 313(3): 600-5, 2004 Jan 16.
Article in English | MEDLINE | ID: mdl-14697233

ABSTRACT

Collagen VI is a main extracellular matrix protein whose mutation is linked to myopathic diseases. In myoblasts and other cell types, collagen VI gene transcription peaks during cell-cycle exit that precedes differentiation, upon serum withdrawal or confluence. To get insight into this transcriptional regulation, we characterized a growth arrest responsive region (GARR) in the Col6a1 promoter responsible for this effect. In this work, we identify sterol regulatory element binding protein (SREBP) as a GARR binding protein and provide evidence that SREBP contributes to induction of Col6a1 transcription in serum free conditions. Furthermore, our data unveil a previously unexpected link between extracellular matrix production and LDL signaling.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Collagen Type VI/biosynthesis , DNA-Binding Proteins/physiology , Transcription Factors , Transcription, Genetic , Animals , Blotting, Northern , Cell Nucleus/metabolism , Collagen Type VI/genetics , Culture Media, Serum-Free/pharmacology , DNA, Complementary/metabolism , Gene Expression Regulation , Gene Library , Glutathione Transferase/metabolism , Lipoproteins, LDL/metabolism , Mice , Muscle Cells/metabolism , NIH 3T3 Cells , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , RNA/metabolism , Sterol Regulatory Element Binding Protein 1 , Transcriptional Activation , Two-Hybrid System Techniques
19.
Nat Genet ; 35(4): 367-71, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625552

ABSTRACT

Collagen VI is an extracellular matrix protein that forms a microfilamentous network in skeletal muscles and other organs. Inherited mutations in genes encoding collagen VI in humans cause two muscle diseases, Bethlem myopathy and Ullrich congenital muscular dystrophy. We previously generated collagen VI-deficient (Col6a1-/-) mice and showed that they have a muscle phenotype that strongly resembles Bethlem myopathy. The pathophysiological defects and mechanisms leading to the myopathic disorder were not known. Here we show that Col6a1-/- muscles have a loss of contractile strength associated with ultrastructural alterations of sarcoplasmic reticulum (SR) and mitochondria and spontaneous apoptosis. We found a latent mitochondrial dysfunction in myofibers of Col6a1-/- mice on incubation with the selective F1F(O)-ATPase inhibitor oligomycin, which caused mitochondrial depolarization, Ca2+ deregulation and increased apoptosis. These defects were reversible, as they could be normalized by plating Col6a1-/- myofibers on collagen VI or by addition of cyclosporin A (CsA), the inhibitor of mitochondrial permeability transition pore (PTP). Treatment of Col6a1-/- mice with CsA rescued the muscle ultrastructural defects and markedly decreased the number of apoptotic nuclei in vivo. These findings indicate that collagen VI myopathies have an unexpected mitochondrial pathogenesis that could be exploited for therapeutic intervention.


Subject(s)
Apoptosis , Collagen Type VI/deficiency , Mitochondria, Muscle/pathology , Mitochondrial Diseases/pathology , Muscular Diseases/pathology , Animals , Calcium/metabolism , Cyclosporine/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/metabolism , Homozygote , Immunosuppressive Agents/pharmacology , In Situ Nick-End Labeling , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Oligomycins/pharmacology , Sarcoplasmic Reticulum/ultrastructure
20.
Biochem Biophys Res Commun ; 309(4): 718-23, 2003 Oct 03.
Article in English | MEDLINE | ID: mdl-13679031

ABSTRACT

HMGA2(2) belongs to the high mobility group A (HMGA) family of architectural transcription factors which participate in a wide variety of nuclear processes ranging from transcription to recombination, playing an important role in chromatin remodelling. HMGA2 is expressed during embryogenesis but not by adult somatic tissues, yet it becomes re-expressed following neoplastic transformation. A role in development is underscored by the finding that the inactivation of the Hmga2 gene is responsible for the murine pygmy phenotype. To elucidate mechanisms that control HMGA2 expression, we have previously cloned the gene and identified functional elements involved in its regulation. In this paper, transgenic mice were generated to define genomic regions involved in Hmga2 developmental and tissue-specific transcriptional regulation. A genomic region from -8.1 to -3.7kb upstream from the initiation site has been found to recapitulate most of the spatial and temporal endogenous Hmga2 gene expression.


Subject(s)
HMGA2 Protein/genetics , Promoter Regions, Genetic , Animals , Gene Expression Regulation, Developmental , Genes, Reporter , In Situ Hybridization , Lac Operon , Mice , Mice, Transgenic , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...