Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters










Publication year range
1.
Biochemistry (Mosc) ; 81(7): 709-22, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27449617

ABSTRACT

In this review we discuss features of mRNA synthesis and modifications used to minimize immune response and prolong efficiency of the translation process in vivo. Considerable attention is given to the use of liposomes and nanoparticles containing lipids and polymers for the mRNA delivery. Finally we briefly discuss mRNAs which are currently in the clinical trials for cancer immunotherapy, vaccination against infectious diseases, and replacement therapy.


Subject(s)
Drug Carriers/chemistry , RNA, Messenger/metabolism , Animals , Humans , Immunity, Innate , Immunotherapy , Liposomes/chemistry , Nanoparticles/chemistry , Neoplasms/immunology , Neoplasms/therapy , RNA, Messenger/chemistry , RNA, Messenger/genetics , Vaccination
2.
Proc Natl Acad Sci U S A ; 98(22): 12521-6, 2001 Oct 23.
Article in English | MEDLINE | ID: mdl-11592978

ABSTRACT

In the vertebrate embryo, neural cell types are organized spatially along the dorsoventral axis of the neural tube and differ by expression of cell-intrinsic determinants and by their adhesive and locomotory properties. Thus, dorsally, neural crest cells (NCC) show a strong propensity to disperse and migrate, whereas cells situated ventrally are highly cohesive and poorly motile. Members of the bone morphogenetic proteins have been shown to exert a dual role in the specification of dorsal neuroepithelial cells and in the dispersion of NCCs. To test whether Sonic hedgehog (Shh), another signaling molecule involved in the patterning of the ventral neural tube, might also contribute to the control of the adhesive and migratory potential of neuroepithelial cells, we analyzed the effect of ectopic Shh on NCC dispersion from neural tube explants cultured in vitro. The addition of Shh to the migration substrate of NCC caused inhibition of their dispersion. The effect of Shh on cell migration was reversible and was not accounted for by alterations of the specification, delamination, proliferation, and survival of NCCs but could be essentially attributed to a decreased cell-substrate adhesion mediated by integrins. In addition, Shh activity on cell migration was mediated by a specific N-terminal region of the molecule and was independent from the signaling cascade elicited by the Patched-Smoothened receptor and involving the Gli transcription factors. Our study therefore reveals an unanticipated role for Shh in regulating adhesion and migration of neuroepithelial cells that is discernable from its inductive, mitogenic, and trophic functions.


Subject(s)
Neural Crest/cytology , Oncogene Proteins/physiology , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Bone Morphogenetic Proteins/physiology , Cell Adhesion , Cell Movement , Chick Embryo , Hedgehog Proteins , Humans , Integrins/physiology , Quail , Smoothened Receptor , Zinc Finger Protein GLI1
3.
J Clin Invest ; 107(12): 1537-44, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11413161

ABSTRACT

We have shown that the integrin alphavbeta6 activates latent TGF-beta in the lungs and skin. We show here that mice lacking this integrin are completely protected from pulmonary edema in a model of bleomycin-induced acute lung injury (ALI). Pharmacologic inhibition of TGF-beta also protected wild-type mice from pulmonary edema induced by bleomycin or Escherichia coli endotoxin. TGF-beta directly increased alveolar epithelial permeability in vitro by a mechanism that involved depletion of intracellular glutathione. These data suggest that integrin-mediated local activation of TGF-beta is critical to the development of pulmonary edema in ALI and that blocking TGF-beta or its activation could be effective treatments for this currently untreatable disorder.


Subject(s)
Antigens, Neoplasm , Respiratory Distress Syndrome/etiology , Transforming Growth Factor beta/physiology , Animals , Bleomycin , Blood-Air Barrier/physiology , Cells, Cultured , Endotoxins , Glutathione/metabolism , Integrins/genetics , Mice , Mice, Knockout , Protein Serine-Threonine Kinases , Pulmonary Alveoli/metabolism , Pulmonary Edema/etiology , Pulmonary Edema/pathology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/administration & dosage , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Transforming Growth Factor beta/antagonists & inhibitors
4.
Gastroenterology ; 119(5): 1286-96, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11054386

ABSTRACT

BACKGROUND & AIMS: Transforming growth factor (TGF)-beta has been implicated in many fibrotic conditions. However, its mechanistic role in radiation toxicity is equivocal despite compelling correlative evidence. This study assessed whether in vivo administration of a soluble TGF-beta type II receptor (TbetaR-II) protein ameliorates intestinal radiation injury (radiation enteropathy). METHODS: A recombinant fusion protein, consisting of the extracellular portion of mouse TbetaR-II and the Fc portion of mouse immunoglobulin (Ig) G, was produced. A 5-cm segment of mouse ileum was exposed to 19 Gy x-radiation. TbetaR-II:Fc fusion protein (1 mg/kg every other day) or mouse IgG was administered from 2 days before to 6 weeks after irradiation. Radiation injury was assessed at 6 weeks using quantitative histology, morphometry, and immunohistochemistry. Collagen was measured colorimetrically, and TGF-beta1 messenger RNA was assessed with fluorogenic probe reverse-transcription polymerase chain reaction. RESULTS: Compared with IgG controls, TbetaR-II:Fc-treated mice exhibited less structural injury, preservation of mucosal surface area, and less intestinal wall fibrosis. Intestinal TGF-beta1 messenger RNA increased in TbetaR-II:Fc-treated mice, whereas TGF-beta immunoreactivity decreased. TbetaR-II:Fc treatment increased crypt cell proliferation but otherwise did not affect unirradiated intestine. CONCLUSIONS: Long-term modulation of TGF-beta with a TbetaR-II:Fc fusion protein is feasible and ameliorates radiation enteropathy. These data confirm the putative role of TGF-beta in intestinal radiation fibrosis.


Subject(s)
Intestinal Diseases/drug therapy , Radiation Injuries/drug therapy , Receptors, Transforming Growth Factor beta/genetics , Recombinant Fusion Proteins/therapeutic use , Animals , CHO Cells , Collagen/metabolism , Cricetinae , Ileum/drug effects , Ileum/metabolism , Ileum/pathology , Ileum/radiation effects , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Intestinal Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/chemistry , Protein Isoforms/genetics , RNA, Messenger/metabolism , Radiation Injuries/pathology , Receptors, Transforming Growth Factor beta/chemistry , Solubility , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
5.
Proc Natl Acad Sci U S A ; 97(13): 7464-9, 2000 Jun 20.
Article in English | MEDLINE | ID: mdl-10861013

ABSTRACT

In the present study, we investigated the role of the CD40L-CD40 pathway in a model of progressive atherosclerosis. ApoE-/- mice were treated with an anti-CD40L antibody or a control antibody for 12 wk. Antibody treatment started early (age 5 wk) or was delayed until after the establishment of atherosclerosis (age 17 wk). In both the early and delayed treatment groups, anti-CD40L antibody did not decrease plaque area or inhibit lesion initiation or age-related increase in lesion area. The morphology of initial lesions was not affected, except for a decrease in T-lymphocyte content. Effects of anti-CD40L antibody treatment on the morphology of advanced lesions were pronounced. In both the early and delayed treatment groups, T-lymphocyte content was significantly decreased. Furthermore, a pronounced increase in collagen content, vascular smooth muscle cell/myofibroblast content, and fibrous cap thickness was observed. In the delayed treatment group, a decrease in lipid core and macrophage content occurred. Interestingly, advanced lesions of anti-CD40L antibody-treated mice exhibited an increased transforming growth factor beta1 immunoreactivity, especially in macrophages. In conclusion, both early and delayed treatment with an anti-CD40L antibody do not affect atherosclerotic lesion initiation but do result in the development of a lipid-poor collagen-rich stable plaque phenotype. Furthermore, delayed treatment with anti-CD40L antibody can transform the lesion profile from a lipid-rich to a lipid-poor collagen-rich phenotype. Postulated mechanisms of this effect on plaque phenotype are the down-regulation of proinflammatory pathways and up-regulation of collagen-promoting factors like transforming growth factor beta.


Subject(s)
Apolipoproteins E/deficiency , Arteriosclerosis , CD40 Antigens/immunology , Membrane Glycoproteins/immunology , Age Factors , Animals , Antibodies/administration & dosage , Antibodies/immunology , Apolipoproteins E/genetics , Arteriosclerosis/genetics , Arteriosclerosis/immunology , CD40 Ligand , Disease Models, Animal , Mice , T-Lymphocytes/immunology
6.
J Clin Invest ; 105(6): 721-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10727440

ABSTRACT

Adhesive interactions play an important role in inflammation by promoting leukocyte attachment and extravasation from the vasculature into the peripheral tissues. However, the importance of adhesion molecules within the extracellular matrix-rich environment of peripheral tissues, in which cells must migrate and be activated, has not been well explored. We investigated the role of the major collagen-binding integrins, alpha1beta1 and alpha2beta1, in several in vivo models of inflammation. mAb's against murine alpha1 and alpha2 were found to significantly inhibit effector phase inflammatory responses in animal models of delayed-type hypersensitivity (DTH), contact hypersensitivity (CHS), and arthritis. Mice that were alpha1-deficient also showed decreased inflammatory responses in the CHS and arthritis models when compared with wild-type mice. Decreased leukocyte infiltration and edema formation accompanied inhibition of antigen-specific models of inflammation, as nonspecific inflammation induced by croton oil was not inhibited. This study demonstrates the importance in vivo of alpha1beta1 and alpha2beta1, the collagen-binding integrins, in inflammatory diseases. The study also extends the role of integrins in inflammation beyond leukocyte attachment and extravasation at the vascular endothelial interface, revealing the extracellular matrix environment of peripheral tissues as a new point of intervention for adhesion-based therapies.


Subject(s)
Arthritis/prevention & control , Cell Adhesion/physiology , Collagen/metabolism , Dermatitis, Allergic Contact/prevention & control , Hypersensitivity, Delayed/prevention & control , Integrins/physiology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Arthritis/immunology , Arthritis/pathology , Collagen/toxicity , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Dermatitis, Irritant/immunology , Dermatitis, Irritant/pathology , Dermatitis, Irritant/prevention & control , Edema/etiology , Edema/prevention & control , Female , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Integrin alpha1beta1 , Integrins/immunology , Leukocytes/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Collagen
7.
Am J Pathol ; 156(1): 115-24, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623659

ABSTRACT

A prominent feature of the hepatic response to injury is production of a fetal isoform of fibronectin, a splice variant containing the EIIIA region, which appears very early after injury and derives from sinusoidal endothelial cells. Previous studies have shown that it is instrumental in initiating the cellular response to injury, specifically the conversion of resting stellate cells to myofibroblast-like cells. The present work describes the regulation of this change in fibronectin expression. Using sinusoidal endothelial cells from normal or injured liver in primary culture, we show that exogenous transforming growth factor beta (TGFbeta) stimulates [EIIIA]Fn expression. To assess the role of TGFbeta in vivo, we used a chimeric IgG containing the extracellular portion of the TGFbeta type II receptor as a competitive inhibitor of the cytokine. Administered to animals at the time of injury, the inhibitor reduced expression of [EIIIA]Fn mRNA by 50% as compared to controls (P < 0.01). There was a corresponding decrease in [EIIIA]Fn protein production as judged by immunohistochemistry. Cell fractionation experiments indicated that the changes observed in whole-liver extracts were localized to sinusoidal endothelial cells. We conclude that TGFbeta initiates wound repair in part by stimulating endothelial expression of [EIIIA]Fn. Results with the soluble inhibitor of the TGFbeta type II receptor suggest a novel strategy for modulating wound repair in vivo.


Subject(s)
Fibronectins/genetics , Genetic Variation , Liver/drug effects , Transforming Growth Factor beta/pharmacology , Wound Healing/physiology , Animals , Autocrine Communication , Bile Ducts , DNA, Recombinant , Endothelium/metabolism , Endothelium/pathology , Ligation , Liver/metabolism , Liver/pathology , Liver/physiology , Male , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta/physiology , Transforming Growth Factor beta/physiology , Up-Regulation
8.
Immunity ; 13(6): 749-58, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11163191

ABSTRACT

Central to immune and inflammatory responses are the integrin-mediated adhesive interactions of cells with their extracellular matrix (ECM)-rich environment. Using a comprehensive and quantitative mRNA profiling technique, we analyzed the effect of ECM-induced attachment on monocyte gene expression, its regulation by growth factors, and the integrin specificity of this event. Adhesion of cells to fibronectin resulted in increased expression of a large number of genes, which was strongly potentiated by the presence of growth factors. Adhesion activated both the NF-kappaB and Jak/STAT pathways of gene transcription and increased expression of genes involved in inflammatory and immune responses, revealing the importance of ECM-integrin interactions in these processes.


Subject(s)
Extracellular Matrix/metabolism , Integrins/metabolism , Monocytes/metabolism , Cell Adhesion/physiology , Cell Line , DNA-Binding Proteins/metabolism , Fibronectins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Growth Substances/metabolism , Humans , Janus Kinase 1 , NF-kappa B/metabolism , Protein-Tyrosine Kinases/metabolism , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/metabolism
9.
Nat Med ; 5(11): 1313-6, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10546000

ABSTRACT

Atherosclerosis is a systemic disease of the large arteries, and activation of inflammatory pathways is important in its pathogenesis. Increasing evidence supports the importance of CD40-CD154 interactions in atherosclerosis, interactions originally known to be essential in major immune reactions and autoimmune diseases. CD40 is present on atheroma-derived cells in vitro and in human atheromata in situ. Ligation of CD40 on atheroma-associated cells in vitro activates the production of chemokines, cytokines, matrix metalloproteinases, adhesion molecules and tissue factor, substances responsible for lesion progression and plaque destabilization. Administration of antibody against CD154 to low-density lipoprotein receptor-deficient mice has been shown to reduce atherosclerosis and decrease T-lymphocyte and macrophage content; however, only initial lesions were studied. Here, we determined the effect of genetic disruption of CD154 in ApoE-/- mice in both initial and advanced atherosclerotic lesions. Plaque area was reduced 550%. In contrast to previous reports, initial lesion development was not affected. Advanced plaques in CD154-/-ApoE-/- mice had a less-lipid-containing, collagen-rich, stable plaque phenotype, with a reduced T-lymphocyte/macrophage content. These data indicate that CD40-CD154 signaling is important in late atherosclerotic changes, such as lipid core formation and plaque destabilization.


Subject(s)
Arteriosclerosis/immunology , Membrane Glycoproteins/immunology , Animals , Base Sequence , CD40 Ligand , Cholesterol/blood , DNA Primers , Disease Progression , Humans , Immunohistochemistry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , Triglycerides/blood
10.
Proc Natl Acad Sci U S A ; 96(22): 12719-24, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10535989

ABSTRACT

Transforming growth factor beta (TGF-beta) is a well characterized cytokine that appears to play a major role in directing the cellular response to injury, driving fibrogenesis, and, thus, potentially underlying the progression of chronic injury to fibrosis. In this study, we report the use of a novel TGF-beta receptor antagonist to block fibrogenesis induced by ligation of the common bile duct in rats. The antagonist consisted of a chimeric IgG containing the extracellular portion of the TGF-beta type II receptor. This "soluble receptor" was infused at the time of injury; in some experiments it was given at 4 days after injury, as a test of its ability to reverse fibrogenesis. The latter was assessed by expression of collagen, both as the mRNA in stellate cells isolated from control or injured liver and also by quantitative histochemistry of tissue sections. When the soluble receptor was administered at the time of injury, collagen I mRNA in stellate cells from the injured liver was 26% of that from animals receiving control IgG (P < 0.0002); when soluble receptor was given after injury induction, collagen I expression was 35% of that in control stellate cells (P < 0.0001). By quantitative histochemistry, hepatic fibrosis in treated animals was 55% of that in controls. We conclude that soluble TGF-beta receptor is an effective inhibitor of experimental fibrogenesis in vivo and merits clinical evaluation as a novel agent for controlling hepatic fibrosis in chronic liver injury.


Subject(s)
Liver Cirrhosis, Experimental/drug therapy , Liver/cytology , Receptors, Transforming Growth Factor beta/physiology , Animals , Collagen/metabolism , Liver Cirrhosis, Experimental/metabolism , Male , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type II , Transforming Growth Factor beta/antagonists & inhibitors
11.
Thorax ; 54(9): 805-12, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10456973

ABSTRACT

BACKGROUND: Transforming growth factor beta (TGF-beta) is a key mediator of collagen synthesis in the development of lung fibrosis. It has previously been shown that the administration of TGF-beta antibody and TGF-beta binding proteoglycan, decorin, reduced bleomycin (BL) induced lung fibrosis in animals. The present study was carried out to investigate whether intratracheal instillation of TGF-beta soluble receptor (TR) would minimise the BL induced lung fibrosis in hamsters. METHODS: The effect of a recombinant TR (TGFbetaRII) on the lung collagen accumulation was evaluated in a BL hamster model of pulmonary fibrosis. Animals were divided into four groups and intratracheally injected with saline or BL at 6.5 U/4 ml/kg followed by intratracheal instillation of phosphate buffered saline (PBS) or 4 nmol TR in 0.3 ml twice a week. Twenty days after the first intratracheal instillation the hamsters were killed for bronchoalveolar lavage (BAL) fluid, biochemical, and histopathological analyses. RESULTS: Treatment of hamsters with TR after intratracheal instillation of BL significantly reduced BL induced lung fibrosis as shown by decreases in the lung hydroxyproline level and prolyl hydroxylase activity, although they were still significantly higher than those of the saline control. Histopathological examination showed a considerable decrease in BL induced fibrotic lesions by TR treatment. However, TR did not prevent the BL induced increases in total cells and protein in the BAL fluid. CONCLUSIONS: These results suggest that TR has antifibrotic potential in vivo and may be useful in the treatment of fibrotic diseases where increased TGF-beta is associated with excess collagen accumulation.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Pulmonary Fibrosis/prevention & control , Receptors, Transforming Growth Factor beta/therapeutic use , Animals , Bronchoalveolar Lavage Fluid , Cell Division/drug effects , Collagen , Cricetinae , Drug Evaluation, Preclinical , Hydroxyproline/pharmacology , Male , Mesocricetus , Mixed Function Oxygenases/pharmacology , Pulmonary Fibrosis/chemically induced , Receptors, Transforming Growth Factor beta/administration & dosage
12.
Circ Res ; 84(10): 1212-22, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10347096

ABSTRACT

Using the rat balloon catheter denudation model, we examined the role of transforming growth factor-beta (TGF-beta) isoforms in vascular repair processes. By en face in situ hybridization, proliferating and quiescent smooth muscle cells in denuded vessels expressed high levels of mRNA for TGF-beta1, TGF-beta2, TGF-beta3, and lower levels of TGF-beta receptor II (TGF-betaRII) mRNA. Compared with normal endothelium, TGF-beta1 and TGF-beta2, as well as TGF-betaRII, mRNA were upregulated in endothelium at the wound edge. Injected recombinant soluble TGF-betaRII (TGF-betaR:Fc) localized preferentially to the adventitia and developing neointima in the injured carotid artery, causing a reduction in intimal lesion formation (up to 65%) and an increase in lumen area (up to 88%). The gain in lumen area was largely due to inhibition of negative remodeling, which coincided with reduced adventitial fibrosis and collagen deposition. Four days after injury, TGF-betaR:Fc treatment almost completely inhibited the induction of smooth muscle alpha-actin expression in adventitial cells. In the vessel wall, TGF-betaR:Fc caused a marked reduction in mRNA levels for collagens type I and III. TGF-betaR:Fc had no effect on endothelial proliferation as determined by reendothelialization of the denuded rat aorta. Together, these findings identify the TGF-beta isoforms as major factors mediating adventitial fibrosis and negative remodeling after vascular injury, a major cause of restenosis after angioplasty.


Subject(s)
Endothelium, Vascular/injuries , Receptors, Transforming Growth Factor beta/metabolism , Actins/metabolism , Angioplasty, Balloon/adverse effects , Animals , Aorta/cytology , Carotid Arteries/chemistry , Carotid Arteries/pathology , Carotid Artery Injuries , Cell Differentiation/physiology , Cell Division/physiology , Collagen/metabolism , Endothelium, Vascular/chemistry , Endothelium, Vascular/pathology , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibroblasts/chemistry , Fibroblasts/cytology , Fibroblasts/enzymology , Fibrosis , Gene Expression/physiology , Hyperplasia , In Situ Hybridization , Ligands , Protein Serine-Threonine Kinases , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Solubility , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tunica Intima/chemistry , Tunica Intima/cytology , Tunica Intima/enzymology
13.
Regul Pept ; 77(1-3): 177-83, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9809813

ABSTRACT

Many extracellular matrix proteins contain the tripeptide sequence arginine-glycine-aspartate (RGD). This RGD motif is recognized by integrins, a family of adhesion receptors present on vascular smooth muscle cells. In the present study, we examined the ability of different RGD-containing peptides to affect the contraction of rat aortic rings in response to different agonists. We found that the peptide RGDS inhibited angiotensin-induced contraction in a dose dependent manner. In contrast, the peptides RGDW and RGES had no effect on angiotensin-induced contractility. We show that function-blocking antibodies to the integrins alphavbeta3 and alpha5beta1 also inhibit angiotensin-induced contraction. These effects were observed in the absence of an intact endothelium. In contrast, neither an antibody directed against the beta1 subunit nor the peptide RGDS had an effect on phenylephrine or 5-hydroxytryptamine-induced contraction. These data suggest that interactions of vascular smooth muscle with components of the surrounding extracellular matrix may influence the response of smooth muscle to agonists.


Subject(s)
Angiotensin II/antagonists & inhibitors , Aorta/drug effects , Integrins/metabolism , Muscle Contraction/drug effects , Oligopeptides/pharmacology , Angiotensin II/pharmacology , Animals , Antibodies/pharmacology , Extracellular Matrix/chemistry , Muscle, Smooth, Vascular/drug effects , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Fibronectin/metabolism , Receptors, Vitronectin/metabolism , Serotonin/pharmacology
14.
Cell Adhes Commun ; 5(3): 193-205, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9686317

ABSTRACT

beta 1D is a recently identified isoform of the beta 1 integrin subunit selectively expressed in skeletal and cardiac muscles. In the present study we determined the temporal expression of beta 1D and its association with alpha subunits during mouse development. By immunohistochemistry and western blot analysis we demonstrated that beta 1D begins to be expressed in skeletal muscles of 17 days embryo (stage E17). Its level progressively increases reaching maximal values few days after birth and remaining high in adult mice. At earlier stages of development (E11-E17) the beta 1A isoform is expressed in skeletal muscle cells. After E17 beta 1A is downregulated and disappears from muscle fibers few days after birth. In cardiac muscle the regulation of the beta 1D expression is different: beta 1D and beta 1A are coexpressed in the heart of E11 embryo. Subsequently expression of beta 1A declines, while beta 1D increases until it becomes the unique beta 1 isoform in cardiomyocytes few days after birth. Previous studies (Belkin et al J. Cell Biol. 132: 211-226, 1996) demonstrated that beta 1D in adult mouse cardiomyocytes is exclusively associated with alpha 7B. Western blot analysis shows that alpha 7B starts to be expressed in the heart only at stage E17, while beta 1D is expressed already at E11 embryo, indicating that alpha subunits other than alpha 7 should associate with beta 1D in early developmental stages. To investigate this aspect, beta 1 associated alpha subunits were identified by western blotting from cardiomyocytes integrin complexes immunoprecipitated with alpha subunit specific antibodies. We found that, during cardiomyocyte development, beta 1D associates with several alpha subunits namely with alpha 5, alpha 6A and alpha 7B. In conclusion these data show that the expression of the beta 1D muscle specific integrin during development occurs much earlier in heart than in skeletal muscle and it can dimerize with different alpha subunits.


Subject(s)
Antigens, CD/genetics , Heart/embryology , Integrin alpha Chains , Integrin beta1/genetics , Muscle, Skeletal/embryology , Alternative Splicing/physiology , Amino Acid Sequence , Animals , Antigens, CD/analysis , Antigens, CD/immunology , Dimerization , Gene Expression Regulation, Developmental/physiology , Immunization , Integrin beta1/analysis , Integrin beta1/immunology , Integrins/analysis , Integrins/genetics , Integrins/immunology , Mice , Molecular Sequence Data , Muscle, Skeletal/chemistry , Myocardium/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology
15.
In Vitro Cell Dev Biol Anim ; 34(3): 217-26, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9557939

ABSTRACT

Transcriptional regulation of smooth muscle cell (SMC) differentiation is a rapidly growing area of interest that has relevance for understanding intimal disease. Despite the wealth of data accumulating in vitro, however, no study has compared the cell-specific marker profile, transfectability, promoter activity, and growth characteristics among several SMC culture systems. Accordingly, we performed a comprehensive analysis of the marker profile, growth properties, transfectability, and SMC promoter activity in four rat SMC lines (A7r5, adult and pup aortic, and PAC1). Despite alterations in chromosomal number and structure, A7r5, adult aortic, and PAC1 cells express all SMC markers studied including SM alpha-actin, SM calponin, SM22, tropoelastin, and to a lesser extent, SM myosin heavy chain (SMMHC). In contrast, pup aortic cells express very low or undetectable levels of all the above markers except tropoelastin. Adult aortic, pup, and PAC1 cells display similar growth curves and levels of proto-oncogene transcripts, whereas those in the A7r5 line are comparatively less. All cell lines studied except pup cells show expression of SMC differentiation genes during active growth, indicating that growth and differentiation are not mutually exclusive in cultured smooth muscle. Transfection studies reveal dramatic differences in DNA uptake and SMC-restricted promoter activity between cell lines. Collectively, these results provide detailed information relating to SMC molecular biology in culture that should facilitate the selection of a cell line for studying the transcriptional regulatory mechanisms underlying SMC differentiation.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Actins/analysis , Animals , Biomarkers , Calcium-Binding Proteins/analysis , Cell Division , Cell Line , Microfilament Proteins , Muscle, Smooth, Vascular/cytology , Myosin Heavy Chains/analysis , Promoter Regions, Genetic , Rats , Rats, Inbred WKY , Rats, Sprague-Dawley , Serum Albumin, Bovine/pharmacology , Transfection , Calponins
16.
Differentiation ; 61(5): 305-11, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9342841

ABSTRACT

Expression of epithelial cytokeratins type 8, 18 and 19 can be used to study smooth muscle cell differentiation during development. We studied the differentiation of smooth muscle cells in the ductus arteriosus before and during intimal thickening and compared the changes occurring in this vessel with the adjoining elastic ascending and descending aorta and the pulmonary trunk. The ductus arteriosus, a vessel connecting the pulmonary trunk and the aorta during fetal life, constricts shorty after birth and eventually closes. Effective closure occurs only in the case of well developed intimal thickening. Cytokeratin expression during fetal development was greatest in the media of the ascending aorta and pulmonary artery, while in the ductus and descending aorta cytokeratin staining was slight. These results suggest that ductus smooth muscle cells and the smooth muscle cells of the descending aorta show a more advanced differentiation as compared to the ascending aorta and pulmonary artery. At neonatal stages cytokeratin expression in the descending aorta, pulmonary artery and the ascending aorta had disappeared as was expected with increased differentiation. In the neonatal ductus arteriosus reexpression of cytokeratins was found in cell clusters in the hyaluronic acid rich environment of the intimal thickening and in the inner media. Reexpression of cytokeratins, especially when organized in clusters, may reflect changes in gene regulation. Therefore the clusters of cytokeratin positive cells in the ductus may be indicative of extensive changes, occurring during closure of this vessel in the neonatal period, in which inner media and intima are especially involved.


Subject(s)
Aorta/metabolism , Ductus Arteriosus/cytology , Keratins/biosynthesis , Muscle, Smooth, Vascular/cytology , Pulmonary Artery/metabolism , Tunica Intima/cytology , Cell Differentiation/physiology , Ductus Arteriosus/anatomy & histology , Ductus Arteriosus/metabolism , Humans , Infant , Infant, Newborn , Muscle Development , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/metabolism , Tunica Intima/anatomy & histology , Tunica Intima/growth & development
17.
Arterioscler Thromb Vasc Biol ; 17(5): 1003-9, 1997 May.
Article in English | MEDLINE | ID: mdl-9157967

ABSTRACT

Differentiation of vascular smooth muscle cells (SMCs) is characterized by several molecular transitions. As differentiation proceeds, proteins of the cytoskeletal and contractile apparatus, such as alpha-smooth muscle actin, smooth muscle myosin, calponin, and heavy caldesmon, and the expression of the membrane-related protein smooth muscle phosphoglucomutase-related protein increase, whereas the expression of other proteins, such as fibronectin splice variants with extradomains A (EDA) and B (EDB), decreases. In this study, we investigated the differentiation of the SMCs of the ductus arteriosus during the development of intimal thickening. Ascending and descending aortas of the same age were used for comparison because these vessels lack intimal thickening. In the fetal ductus arteriosus, a relatively early differentiation of the contractile apparatus was observed compared with the ascending and descending aortas. EDA and EDB expression was already low, being similar in the ductus and descending aorta and even lower in the ascending aorta. In the neonatal ductus, SMCs of the media and outer intima were well differentiated and comparable with SMCs of the ascending aorta. Dedifferentiated SMCs, with a low expression of cytoskeletal and contractile proteins and a high expression of EDA and EDB, were found in regions in the inner intima that show features of progression of intimal thickening and in areas of cytolytic necrosis in the media. With a technique using in situ end labeling of DNA fragments, we found extensive apoptosis in the area of cytolytic necrosis and to a lesser extent in these areas of the inner intima. In conclusion, SMCs of the fetal ductus arteriosus have an advanced differentiation of the contractile apparatus compared with the adjacent aorta. Reexpression of fetal characteristics is seen in a number of cells in inner intima and media of the neonatal ductus arteriosus. The finding of apoptosis in these areas suggests that dedifferentiation and apoptosis are associated processes that may play a role in vascular remodeling.


Subject(s)
Apoptosis , Cell Differentiation , Ductus Arteriosus/embryology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/embryology , Aorta/cytology , Aorta/embryology , Aorta/growth & development , DNA Fragmentation , Ductus Arteriosus/cytology , Ductus Arteriosus/growth & development , Fetus/blood supply , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , In Situ Hybridization , Infant, Newborn , Muscle Development , Muscle, Smooth, Vascular/growth & development
18.
J Biol Chem ; 272(50): 31447-52, 1997 Dec 12.
Article in English | MEDLINE | ID: mdl-9395478

ABSTRACT

Many cell-surface and extracellular matrix proteins contain multiple modular domains known as fibronectin type III (FNIII) repeats. Cells adhere to the extracellular matrix proteins fibronectin and tenascin in part by the interaction of certain integrins with the Arg-Gly-Asp (RGD) sequence, displayed on specific FNIII repeats. We have found that, after experimental activation of beta1 integrins, a number of cell types adhere and spread on FNIII repeats lacking RGD, derived from extracellular matrix proteins and cytokine receptors. Interaction between individual FNIII domains and beta1 integrins mediates focal adhesion kinase phosphorylation and subsequent stress fiber and focal contact formation. These data suggest that many FNIII-containing proteins may bind and signal through activated beta1 integrins, dramatically expanding the potential for integrin-dependent intercellular and cell-matrix communication.


Subject(s)
Cell Adhesion/physiology , Fibronectins/chemistry , Integrin beta1/physiology , Oligopeptides/physiology , Signal Transduction/physiology , Animals , Binding Sites , Calcium/metabolism , Cell Adhesion Molecules/metabolism , Cell Communication/physiology , Extracellular Matrix Proteins/chemistry , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Manganese/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Receptors, Cytokine/chemistry
19.
J Cell Biol ; 139(6): 1583-95, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9396762

ABSTRACT

Expression of muscle-specific beta1D integrin with an alternatively spliced cytoplasmic domain in CHO and GD25, beta1 integrin-minus cells leads to their phenotypic conversion. beta1D-transfected nonmuscle cells display rounded morphology, lack of pseudopodial activity, retarded spreading, reduced migration, and significantly enhanced contractility compared with their beta1A-expressing counterparts. The transfected beta1D is targeted to focal adhesions and efficiently displaces the endogenous beta1A and alphavbeta3 integrins from the sites of cell-matrix contact. This displacement is observed on several types of extracellular matrix substrata and leads to elevated stability of focal adhesions in beta1D transfectants. Whereas a significant part of cellular beta1A integrin is extractable in digitonin, the majority of the transfected beta1D is digitonin-insoluble and is strongly associated with the detergent-insoluble cytoskeleton. Increased interaction of beta1D integrin with the actin cytoskeleton is consistent with and might be mediated by its enhanced binding to talin. In contrast, beta1A interacts more strongly with alpha-actinin, than beta1D. Inside-out driven activation of the beta1D ectodomain increases ligand binding and fibronectin matrix assembly by beta1D transfectants. Phenotypic effects of beta1D integrin expression in nonmuscle cells are due to its enhanced interactions with both cytoskeletal and extracellular ligands. They parallel the transitions that muscle cells undergo during differentiation. Modulation of beta1 integrin adhesive function by alternative splicing serves as a physiological mechanism reinforcing the cytoskeleton- matrix link in muscle cells. This reflects the major role for beta1D integrin in muscle, where extremely stable association is required for contraction.


Subject(s)
Alternative Splicing , Cell Adhesion , Cytoskeleton/physiology , Extracellular Matrix/physiology , Integrin beta1/physiology , Muscles/physiology , Actins/physiology , Actins/ultrastructure , Animals , CHO Cells , Cell Line , Cricetinae , Cytoskeleton/ultrastructure , DNA, Complementary , Extracellular Matrix/ultrastructure , Humans , Integrin beta1/biosynthesis , Muscle Contraction , Myosin Light Chains/metabolism , Phosphorylation , Receptors, Vitronectin/physiology , Recombinant Fusion Proteins/biosynthesis , Transfection
20.
J Clin Invest ; 97(11): 2469-77, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8647938

ABSTRACT

Remodeling of the extracellular matrix by activated mesenchymal cells (myofibroblasts) is a critical aspect of wound repair in all adult organs. Collagen-dependent gel contraction, a process requiring integrin function, is an established in vitro assay thought to mimic in vivo matrix remodeling. Numerous data have implicated the alpha2beta1 integrin in various cell types as the primary collagen receptor responsible for collagen gel contraction. However, evidence from the literature suggests that the major collagen binding integrin expressed on mesenchymally derived cells in situ is the alpha1beta1 integrin, not the alpha2beta1 integrin. In this report, we use a rat vascular injury model to illustrate that the alpha1beta1 integrin is the major collagen receptor expressed on vascular smooth muscle cells after injury. Using two smooth muscle cell lines, expressing either the alpha1beta1 integrin alone or both the alpha1beta1 and alpha2beta1 integrins, along with Chinese hamster ovary cells transfected with the alpha1 integrin, we demonstrate that alpha1beta1 supports not only collagen-dependent adhesion and migration, but also gel contraction. These data suggest that in vivo the alpha1beta1 integrin is a critical collagen receptor on mesenchymally derived cells potentially involved in matrix remodeling after injury.


Subject(s)
Carotid Artery, Common/physiology , Collagen/metabolism , Integrins/biosynthesis , Muscle, Smooth, Vascular/physiology , Tunica Intima/physiology , Wound Healing , Animals , Antigens, CD/biosynthesis , Aorta/injuries , Aorta/physiology , CHO Cells , Carotid Artery Injuries , Cell Adhesion , Cell Line , Cell Movement , Cricetinae , Extracellular Matrix/physiology , Humans , Immunohistochemistry , In Situ Hybridization , Integrin alpha1 , Integrin alpha1beta1 , Male , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...