Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Diabetes Metab ; 44(3): 292-295, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29129540

ABSTRACT

AIM: Oxytocin administration to diet-induced obese (DIO) rodents, monkeys and humans decreases body weight and fat mass with concomitant improvements in glucose metabolism. Moreover, several studies show an immunomodulatory role of oxytocin in a number of settings (such as atherosclerosis, injury, sepsis). This study aims to shed some light on the effects of oxytocin on macrophage polarization and cytokine production, as well as its possible impact on these parameters in adipose tissue in DIO mice with impaired glucose metabolism. METHODS: Mouse bone marrow cells were differentiated into macrophages and treated with oxytocin. Macrophage proliferation, cytokine secretion and macrophage populations were determined. For experiments in vivo, DIO mice were treated with oxytocin for 2 weeks. Body weight and composition and glucose tolerance were subsequently followed. At the end of treatment, adipose tissue macrophage populations, plasma cytokine levels and cytokine expression in adipose tissue were determined. RESULTS: In bone marrow-derived macrophages, oxytocin induced an anti-inflammatory phenotype (decreased M1/M2 ratio). In M1-derived macrophages, oxytocin decreased TNFα secretion, with no effects on the other cytokines tested nor any effect on cytokine secretion by M2-derived macrophages. Oxytocin treatment in DIO mice in vivo led to decreased body weight accompanied by an improvement in glucose tolerance, with no changes in plasma cytokine levels. In adipose tissue, oxytocin decreased Tnfα expression without modifying the M1/M2 macrophage ratio. CONCLUSION: Oxytocin treatment decreases TNFα production both in vitro (in bone marrow-derived macrophages) and in vivo (in epididymal adipose tissue) in DIO mice. This effect may also be contributory to the observed improvement in glucose metabolism.


Subject(s)
Adipose Tissue/drug effects , Macrophages/drug effects , Oxytocin/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Adipose Tissue/metabolism , Animals , Body Composition/drug effects , Body Weight/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Macrophages/metabolism , Mice
2.
J Cell Biol ; 142(3): 873-81, 1998 Aug 10.
Article in English | MEDLINE | ID: mdl-9700173

ABSTRACT

Transforming growth factor-beta1 (TGFbeta1), a major promoter of myofibroblast differentiation, induces alpha-smooth muscle (sn) actin, modulates the expression of adhesive receptors, and enhances the synthesis of extracellular matrix (ECM) molecules including ED-A fibronectin (FN), an isoform de novo expressed during wound healing and fibrotic changes. We report here that ED-A FN deposition precedes alpha-SM actin expression by fibroblasts during granulation tissue evolution in vivo and after TGFbeta1 stimulation in vitro. Moreover, there is a correlation between in vitro expression of alpha-SM actin and ED-A FN in different fibroblastic populations. Seeding fibroblasts on ED-A FN does not elicit per se alpha-SM actin expression; however, incubation of fibroblasts with the anti-ED-A monoclonal antibody IST-9 specifically blocks the TGFbeta1-triggered enhancement of alpha-SM actin and collagen type I, but not that of plasminogen activator inhibitor-1 mRNA. Interestingly, the same inhibiting action is exerted by the soluble recombinant domain ED-A, but neither of these inhibitory agents alter FN matrix assembly. Our findings indicate that ED-A-containing polymerized FN is necessary for the induction of the myofibroblastic phenotype by TGFbeta1 and identify a hitherto unknown mechanism of cytokine-determined gene stimulation based on the generation of an ECM-derived permissive outside in signaling, under the control of the cytokine itself.


Subject(s)
Cell Differentiation/physiology , Fibroblasts/cytology , Fibronectins/physiology , Muscle, Smooth/cytology , Transforming Growth Factor beta/physiology , Actins/metabolism , Animals , Extracellular Matrix/physiology , Female , Fibronectins/chemistry , Humans , Phenotype , Rats , Rats, Wistar , Signal Transduction
3.
Am J Pathol ; 150(2): 509-21, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9033267

ABSTRACT

Previous work (M.-L. Bochaton-Piallat, P. Ropraz, F. Gabbiani, G. Gabbiani, Arterioscler Thromb Vasc Biol 1996, 16:815-820) has shown that a subset of smooth muscle cell (SMC) clones derived from the normal rat aortic media displays an epithelioid phenotype similar to that of the whole SMC population cultured from the intimal thickening 15 days after endothelial injury (IT-15). We show here that the whole IT-15 SMC population and the epithelioid clones, derived either from the normal media or from the IT-15, express cellular retinol-binding protein-1 (CRBP-1), a protein involved in retinoid metabolism. The expression of CRBP-1 is accompanied by the expression of cytokeratin 8. In both whole SMC population cultured from IT-15 and epithelioid clones, retinoic acid modulates the transition from the epithelioid phenotype to the spindle phenotype, typical of whole SMC populations cultured from the rat normal aortic media. Moreover, after endothelial injury in vivo, a CRBP-1 expressing SMC subset appears transiently in the IT and disappears, allegedly by apoptosis, when re-endothelialization takes place. Our results suggest that the expression of CRBP-1 is a marker of arterial SMC activation after endothelial injury in vivo and that CRBP-1 and probably retinoids participate in this process.


Subject(s)
Aorta/metabolism , Muscle, Smooth, Vascular/metabolism , Retinol-Binding Proteins/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Clone Cells , Keratins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Retinol-Binding Proteins/genetics , Retinol-Binding Proteins, Cellular , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Vitamin A/pharmacology
4.
Arterioscler Thromb Vasc Biol ; 16(6): 815-20, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8640410

ABSTRACT

It is well accepted that smooth muscle cells (SMCs) cultured from normal rat arterial media have different morphological and biological features compared with SMCs cultured from experimental intimal thickening (IT) 15 days after endothelial injury. It is not known, however, whether the phenotypic modulation producing IT cells occurs in any medial SMCs or only in a particular SMC subpopulation. To distinguish among these possibilities, the phenotypic features of SMC clones derived from normal adult media and the IT 15 days after endothelial lesion were analyzed according to morphological appearance, replicative activity in the presence and absence of fetal calf serum, and [3H]thymidine incorporation and motile activity; these features were compared with those of the respective SMC parental populations. Two categories of SMC clones predominated: spindle clones, with morphological features similar to those of the parental population from the normal media, and epithelioid clones, with morphological features similar to those of the IT parental population. Both categories were present among clones produced from normal media and IT; however, spindle was more common among normal media clones, and epithelioid, among IT clones. The behavior in vitro was distinct for each category of clones and did not depend on their origin. Our results are compatible with the possibility that the SMC population of IT in vivo derives mainly from SMCs belonging to the category exhibiting epithelioid features in vitro.


Subject(s)
Aorta, Thoracic/injuries , Endothelium, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Animals , Aorta, Thoracic/pathology , Cell Division , Cell Movement , Clone Cells/ultrastructure , Cytoskeleton/ultrastructure , DNA Replication , Embolectomy/adverse effects , Phenotype , Rats , Rats, Wistar
5.
Exp Cell Res ; 214(2): 528-36, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7925646

ABSTRACT

Locally liberated cytokines and extracellular matrix components influence the proliferation and differentiation of arterial smooth muscle cells (SMC), thus playing a role in the development of the atheromatous plaque. It has been proposed that the response of SMC to these factors is influenced by their own phenotype. We have tested the effects of transforming growth factor-beta 1 (TGF-beta 1) and heparin on proliferation and expression of alpha-smooth muscle (SM) actin, a well-established SMC differentiation marker, by cultured rat SMC obtained from the normal aorta of young or old rats and from the intimal thickening developed 15 days after endothelial denudation in young rats; these SMC are known to express different phenotypic features. Heparin and TGF-beta 1 reduced serum-induced proliferation in SMC from young and old rats. Heparin increased the expression of alpha-SM actin protein and mRNA in SMC from young and old rats, while TGF-beta 1 exerted the opposite action. Moreover, TGF-beta 1 induced the appearance of an elongated shape in SMC from both young and old rats. In SMC cultured from intimal thickening, heparin induced a reduction of cell proliferation without modifying their characteristic epithelioid shape; TGF-beta 1 increased the proliferative activity and induced an elongated cell shape as well as a "hills and valleys" growth pattern similar to that observed in control medial SMC; both heparin and TGF-beta 1 induced an increase of alpha-SM actin expression. Our results show that TGF-beta 1 and heparin exert different effects on the same SMC, suggesting that these substances act at least in part independently. They are also compatible with the view that the action of cytokines and of extracellular matrix components depends on the phenotype of target SMC.


Subject(s)
Actins/biosynthesis , Aorta/cytology , Heparin/pharmacology , Muscle, Smooth, Vascular/physiology , Transforming Growth Factor beta/pharmacology , Actins/genetics , Age Factors , Animals , Cell Division/drug effects , Cell Size/drug effects , Cytoskeleton/physiology , Dose-Response Relationship, Drug , Female , Muscle, Smooth, Vascular/drug effects , RNA, Messenger/analysis , Rats , Rats, Wistar , Tunica Intima/cytology
6.
Arterioscler Thromb ; 14(6): 982-9, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8199190

ABSTRACT

Endothelial denudation by balloon injury of the rat aorta induces the development of a neointima as a consequence of the migration and proliferation of smooth muscle cells (SMCs). Initially, intimal SMCs show a dedifferentiated phenotype, which reverts to a normal differentiated phenotype after endothelial cells have resurfaced the vessel lumen. We investigated in vitro the proliferative and phenotypic features of SMCs from different layers of rat aorta isolated 15 and 60 days after endothelial denudation. Freshly isolated intimal cells 15 days after balloon injury (IT-15) appeared rounded and showed a decreased content of alpha-smooth muscle actin, smooth muscle myosin, and desmin compared with intimal cells isolated 60 days after balloon injury (IT-60). No morphological and cytoskeletal differences were observed among freshly isolated IT-60 cells and other medial populations, which included medial SMCs that underlie the intimal thickening. In culture, IT-15 cells showed increased proliferative activity both in monolayers and in free-floating collagen lattices. Decreased expression of alpha-smooth muscle actin and smooth muscle myosin was documented in IT-15 cells compared with IT-60 cells and other medial SMC populations in monolayer. Moreover, IT-15 cells suspended in collagen lattices were poor at contracting these collagen lattices compared with IT-60 and control SMCs. IT-60 cells were equivalent to control SMCs at lattice contraction except for a temporary delay at day 1.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Muscle, Smooth, Vascular/cytology , Actins/analysis , Actins/genetics , Animals , Aorta, Thoracic/cytology , Cell Division , Cells, Cultured/chemistry , Cells, Cultured/cytology , Collagen/physiology , Cytoskeleton/chemistry , Endothelium, Vascular/physiology , Isomerism , Male , Muscle Contraction , Myosins/analysis , RNA, Messenger/analysis , Rats , Rats, Wistar
7.
Arterioscler Thromb ; 13(10): 1449-55, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8399081

ABSTRACT

The replicative activity and the differentiation features of aortic smooth muscle cells (SMCs) cultured as whole populations or clones from newborn (4-day-old), young adult (6-week-old), and old (18-month-old) rats were studied by means of cell counting, [3H]thymidine incorporation, and measurement of the expression of cytoskeletal proteins and mRNAs. In whole populations at the fifth passage, replicative activity increased and differentiation features (ie, expression of alpha-smooth muscle actin, desmin, and smooth muscle myosin heavy chains) decreased with increasing age of the donor animal. SMC clones derived from newborn or young adult rats showed more differentiated cytoskeletal features than their parental populations; however, most SMC clones from old rats showed dedifferentiated features similar to those observed in their parental populations. Our results suggest that (1) SMCs of the rat aortic media behave as a heterogeneous population; (2) cultured whole SMC populations behave differently from clones as far as their replicative activity and differentiation features are concerned; and (3) SMCs derived from old rats, whether grown as whole populations or as clones, dedifferentiate more substantially and replicate more actively than corresponding cultures from newborn or young adult rats when submitted to the same amount of serum growth factors; these differences may play a role in arterial development as well as in the formation and evolution of the atheromatous plaque.


Subject(s)
Aging/physiology , Aorta/cytology , Muscle, Smooth, Vascular/cytology , Actins/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Cell Differentiation , Cell Division , Cells, Cultured , Clone Cells , Desmin/metabolism , Isomerism , Muscle Development , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/metabolism , Myosins/metabolism , Rats , Rats, Wistar
8.
Differentiation ; 49(3): 175-85, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1377654

ABSTRACT

It is well known that arterial smooth muscle cells (SMC) of adult rats, cultured in a medium containing fetal calf serum (FCS), replicate actively and lose the expression of differentiation markers, such as desmin, smooth muscle (SM) myosin and alpha-SM actin. We report here that compared to freshly isolated cells, primary cultures of SMC from newborn animals show no change in the number of alpha-SM actin containing cells and a less important decrease in the number of desmin and SM myosin containing cells than that seen in primary cultures of SMC from adult animals; moreover, contrary to what is seen in SMC cultured from adult animals, they show an increase of alpha-SM actin mRNA level, alpha-SM actin synthesis and expression per cell. These features are partially maintained at the 5th passage, when the cytoskeletal equipment of adult SMC has further evolved toward dedifferentiation. Cloned newborn rat SMC continue to express alpha-SM actin, desmin and SM myosin at the 5th passage. Thus, newborn SMC maintain, at least in part, the potential to express differentiated features in culture. Heparin has been proposed to control proliferation and differentiation of arterial SMC. When cultured in the presence of heparin, newborn SMC show an increase of alpha-SM actin synthesis and content but no modification of the proportion of alpha-SM actin total (measured by Northern blots) and functional (measured by in vitro translation in a reticulocyte lysate) mRNAs compared to control cells cultured for the same time in FCS containing medium. This suggests that heparin action is exerted at a translational or post-translational level. Cultured newborn rat aortic SMC furnish an in vitro model for the study of several aspects of SMC differentiation and possibly of mechanisms leading to the establishment and prevention of atheromatous plaques.


Subject(s)
Aorta, Thoracic/growth & development , Cytoskeletal Proteins/metabolism , Cytoskeleton/ultrastructure , Muscle Development , Muscle, Smooth, Vascular/growth & development , Actins/metabolism , Aging , Animals , Animals, Newborn , Aorta, Thoracic/metabolism , Aorta, Thoracic/ultrastructure , Blotting, Northern , Cells, Cultured , Clone Cells , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Desmin/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/ultrastructure , Myosins/metabolism , RNA/genetics , RNA/isolation & purification , Rats , Rats, Inbred Strains
9.
J Cell Biol ; 107(5): 1939-45, 1988 Nov.
Article in English | MEDLINE | ID: mdl-2460473

ABSTRACT

Quiescent smooth muscle cells (SMC) in normal artery express a pattern of actin isoforms with alpha-smooth muscle (alpha SM) predominance that switches to beta predominance when the cells are proliferating. We have examined the relationship between the change in actin isoforms and entry of SMC into the growth cycle in an in vivo model of SMC proliferation (balloon injured rat carotid artery). alpha SM actin mRNA declined and cytoplasmic (beta + gamma) actin mRNAs increased in early G0/G1 (between 1 and 8 h after injury). In vivo synthesis and in vitro translation experiments demonstrated that functional alpha SM mRNA is decreased 24 h after injury and is proportional to the amount of mRNA present. At 36 h after injury, SMC prepared by enzymatic digestion were sorted into G0/G1 and S/G2 populations; only the SMC committed to proliferate (S/G2 fraction) showed a relative slight decrease in alpha SM actin and, more importantly, a large decrease in alpha SM actin mRNA. A switch from alpha SM predominance to beta predominance was present in the whole SMC population 5 d after injury. To determine if the change in actin isoforms was associated with proliferation, we inhibited SMC proliferation by approximately 80% with heparin, which has previously been shown to block SMC in late G0/G1 and to reduce the growth fraction. The switch in actin mRNAs and synthesis at 24 h was not prevented; however, alpha SM mRNA and protein were reinduced at 5 d in the heparin-treated animals compared to saline-treated controls. These results suggest that in vivo the synthesis of actin isoforms in arterial SMC depends on the mRNA levels and changes after injury in early G0/G1 whether or not the cells subsequently proliferate. The early changes in actin isoforms are not prevented by heparin, but they are eventually reversed if the SMC are kept in the resting state by the heparin treatment.


Subject(s)
Actins/biosynthesis , Heparin/pharmacology , Muscle, Smooth, Vascular/cytology , Actins/genetics , Animals , Blotting, Northern , Cell Cycle , Cell Division/drug effects , Cell Separation , Electrophoresis, Polyacrylamide Gel , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , RNA/isolation & purification , Rats , Rats, Inbred Strains
10.
J Cell Biol ; 103(6 Pt 2): 2787-96, 1986 Dec.
Article in English | MEDLINE | ID: mdl-3539945

ABSTRACT

A monoclonal antibody (anti-alpha sm-1) recognizing exclusively alpha-smooth muscle actin was selected and characterized after immunization of BALB/c mice with the NH2-terminal synthetic decapeptide of alpha-smooth muscle actin coupled to keyhole limpet hemocyanin. Anti-alpha sm-1 helped in distinguishing smooth muscle cells from fibroblasts in mixed cultures such as rat dermal fibroblasts and chicken embryo fibroblasts. In the aortic media, it recognized a hitherto unknown population of cells negative for alpha-smooth muscle actin and for desmin. In 5-d-old rats, this population is about half of the medial cells and becomes only 8 +/- 5% in 6-wk-old animals. In cultures of rat aortic media SMCs, there is a progressive increase of this cell population together with a progressive decrease in the number of alpha-smooth muscle actin-containing stress fibers per cell. Double immunofluorescent studies carried out with anti-alpha sm-1 and anti-desmin antibodies in several organs revealed a heterogeneity of stromal cells. Desmin-negative, alpha-smooth muscle actin-positive cells were found in the rat intestinal muscularis mucosae and in the dermis around hair follicles. Moreover, desmin-positive, alpha-smooth muscle actin-negative cells were identified in the intestinal submucosa, rat testis interstitium, and uterine stroma. alpha-Smooth muscle actin was also found in myoepithelial cells of mammary and salivary glands, which are known to express cytokeratins. Finally, alpha-smooth muscle actin is present in stromal cells of mammary carcinomas, previously considered fibroblastic in nature. Thus, anti-alpha sm-1 antibody appears to be a powerful probe in the study of smooth muscle differentiation in normal and pathological conditions.


Subject(s)
Actins/immunology , Antibodies, Monoclonal/immunology , Muscle, Smooth/immunology , Actins/metabolism , Animals , Antibody Specificity , Aorta/cytology , Desmin/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Mammary Glands, Animal/metabolism , Muscle, Smooth/metabolism , Muscles/metabolism , Myocardium/metabolism , Salivary Glands/metabolism
11.
J Submicrosc Cytol ; 18(3): 481-93, 1986 Jul.
Article in English | MEDLINE | ID: mdl-3528513

ABSTRACT

Cytoskeletal features of arterial smooth muscle cells (SMC) vary characteristically during development and during atheromatous plaque formation (Gabbiani et al., 1984; Kocher et al., 1985). We have analyzed the cytoskeletal features of rat aortic SMC placed in culture in the presence of 10% foetal calf serum (thus containing growth factors probably playing a role in SMC development and atheroma formation), as compared to SMC freshly isolated from the rat aortic media. Under these conditions, SMC show a typical cytoskeletal remodeling characterized by: 1) increased content of vimentin per cell, increased number of cells containing only vimentin, and decreased number of vimentin plus desmin containing cells; 2) decreased contents of actin, tropomyosin and myosin; 3) a switch in the pattern of actin isoforms with the appearance of a beta-type predominance. Some of these changes (e.g. increase of vimentin and decrease of alpha-type actin) are seen already in cells entering for the first time in S-phase after plating. Pulse-chase experiments with 3H-thymidine (3H-TdR) indicate that vimentin containing SMC possess a higher replicative activity than vimentin plus desmin containing SMC, thus explaining the selection of vimentin containing cells during culture. Our results indicate that during culture SMC develop features similar to those observed in normal foetal SMC or in SMC present in atheromatous plaques; this model may be useful for the understanding of mechanisms leading to SMC differentiation and to atheroma formation.


Subject(s)
Cytoskeletal Proteins/analysis , Cytoskeleton/ultrastructure , Muscle, Smooth, Vascular/cytology , Animals , Aorta/cytology , Aorta/ultrastructure , Cell Division/drug effects , Cells, Cultured , DNA Replication , Desmin/analysis , Electrophoresis, Polyacrylamide Gel , Fibrin/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Muscle, Smooth, Vascular/ultrastructure , Rats , Rats, Inbred Strains , Vimentin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...