Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Wound Repair Regen ; 21(1): 166-76, 2013.
Article in English | MEDLINE | ID: mdl-23253249

ABSTRACT

The contractile phenotype and function of myofibroblasts have been proposed to play a critical role in wound closure. It has been hypothesized that smooth muscle α-actin expressed in myofibroblasts is critical for its formation and function. We have used smooth muscle α-actin-null mice to test this hypothesis. Full-thickness excisional wounds closed at a similar rate in smooth muscle α-actin-null and wild-type mice. In addition, fibroblasts in smooth muscle α-actin-null granulation tissue when immunostained with a monoclonal antibody that recognizes all muscle actin isoforms exhibited a myofibroblast-like distribution and a stress fiber-like pattern, showing that these cells acquired the myofibroblast phenotype. Dermal fibroblasts from smooth muscle α-actin-null and wild-type mice formed stress fibers and supermature focal adhesions, and generated similar amounts of contractile force in response to transforming growth factor-ß1. Smooth muscle γ-actin and skeletal muscle α-actin were expressed in smooth muscle α-actin-null myofibroblasts, as shown by immunostaining, real-time polymerase chain reaction, and mass spectrometry. These results show that smooth muscle α-actin is not necessary for myofibroblast formation and function and for wound closure, and that smooth muscle γ-actin and skeletal muscle α-actin may be able to functionally compensate for the lack of smooth muscle α-actin in myofibroblasts.


Subject(s)
Actins/metabolism , Fibroblasts/pathology , Focal Adhesions/pathology , Granulation Tissue/pathology , Myofibroblasts/pathology , Wound Healing , Wounds and Injuries/pathology , Animals , Blotting, Western , Cell Differentiation , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
2.
Exp Cell Res ; 318(13): 1542-53, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22449415

ABSTRACT

During wound healing, fibroblasts transition from quiescence to a migratory state, then to a contractile myofibroblast state associated with wound closure. We found that the myofibroblast phenotype, characterized by the expression of high levels of contractile proteins, suppresses the expression of the pro-migratory gene, MMP-2. Fibroblasts cultured in a 3-D collagen lattice and allowed to develop tension showed increased contractile protein expression and decreased MMP-2 levels in comparison to a stress-released lattice. In 2-D cultures, factors that promote fibroblast contractility, including serum or TGF-ß, down-regulated MMP-2. Pharmacologically inducing F-actin disassembly or reduced contractility increased MMP-2 expression, while conditions that promote F-actin assembly suppressed MMP-2 expression. In all cases, changes in MMP-2 levels were inversely related to changes in the contractile marker, smooth muscle α-actin. To determine if the mechanisms involved in contractile protein gene expression play a direct role in MMP-2 regulation, we used RNAi-mediated knock-down of the myocardin-like factors, MRTF-A and MRTF-B, which induced the down-regulation of contractile protein genes by fibroblasts under both serum-containing and serum-free conditions. In the presence of serum or TGF-ß, MRTF-A/B knock-down resulted in the up-regulation of MMP-2; serum-free conditions prevented this increased expression. Together, these results indicate that, while MMP-2 expression is suppressed by F-actin formation, its up-regulation is not simply a consequence of contractile protein down-regulation.


Subject(s)
Fibroblasts/enzymology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Myofibroblasts/enzymology , Actins/chemistry , Actins/metabolism , Animals , Cell Culture Techniques/methods , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Insulin-Like Growth Factor I/pharmacology , Models, Biological , Myofibroblasts/cytology , Myofibroblasts/physiology , Phenotype , Protein Multimerization , RNA Interference , Rats , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Wound Healing/genetics , Wound Healing/physiology
3.
J Invest Dermatol ; 131(12): 2378-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21776010

ABSTRACT

Myofibroblasts are contractile, smooth muscle-like cells that are characterized by the de novo expression of smooth muscle α-actin (SMαA) and normally function to assist in wound closure, but have been implicated in pathological contractures. Transforming growth factor ß-1 (TGF-ß1) helps facilitate the differentiation of fibroblasts into myofibroblasts, but the exact mechanism by which this differentiation occurs, in response to TGF-ß1, remains unclear. Myocardin-related transcription factors A and B (MRTFs, MRTF-A/B) are transcriptional co-activators that regulate the expression of smooth muscle-specific cytoskeletal proteins, including SMαA, in smooth muscle cells and fibroblasts. In this study, we demonstrate that TGF-ß1 mediates myofibroblast differentiation and the expression of a contractile gene program through the actions of the MRTFs. Transient transfection of a constitutively active MRTF-A induced an increase in the expression of SMαA and other smooth muscle-specific cytoskeletal proteins, and an increase in myofibroblast contractility, even in the absence of TGF-ß1. MRTF-A/B knockdown, in TGF-ß1-differentiated myofibroblasts, resulted in decreased smooth muscle-specific cytoskeletal protein expression levels and reduced contractile force generation, as well as a decrease in focal adhesion size and number. These results provide direct evidence that the MRTFs are mediators of myofibroblast differentiation in response to TGF-ß1.


Subject(s)
Cell Differentiation/genetics , Fibroblasts/cytology , Myofibroblasts/cytology , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Animals , Cell Line , Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , Focal Adhesions/metabolism , Myofibroblasts/metabolism , Rats
4.
Biol Reprod ; 85(1): 13-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21368298

ABSTRACT

Mammary myoepithelial cells are specialized smooth musclelike epithelial cells that express the smooth muscle actin isoform: smooth muscle alpha-actin (ACTA2). These cells contract in response to oxytocin to generate the contractile force required for milk ejection during lactation. It is believed that ACTA2 contributes to myoepithelial contractile force generation; however, this hypothesis has not been directly tested. To evaluate the contribution of ACTA2 to mammary myoepithelial cell contraction, Acta2 null mice were utilized and milk ejection and myoepithelial cell contractile force generation were evaluated. Pups suckling on Acta2 null dams had a significant reduction in weight gain starting immediately postbirth. Cross-fostering demonstrated the lactation defect is with the Acta2 null dams. Carmine alum whole mounts and conventional histology revealed no underlying structural defects in Acta2 null mammary glands that could account for the lactation defect. In addition, myoepithelial cell formation and organization appeared normal in Acta2 null lactating mammary glands as evaluated using an Acta2 promoter-GFP transgene or phalloidin staining to visualize myoepithelial cells. However, mammary myoepithelial cell contraction in response to oxytocin was significantly reduced in isolated Acta2 null lactating mammary glands and in in vivo studies using Acta2 null lactating dams. These results demonstrate that lack of ACTA2 expression impairs mammary myoepithelial cell contraction and milk ejection and suggests that ACTA2 expression in mammary myoepithelial cells has the functional consequence of enhancing contractile force generation required for milk ejection.


Subject(s)
Actins/metabolism , Mammary Glands, Animal/physiology , Milk Ejection , Animals , Animals, Newborn , Failure to Thrive , Female , Mammary Glands, Animal/cytology , Mice , Mice, Knockout , Oxytocin/physiology , Pregnancy
5.
Wound Repair Regen ; 18(2): 223-34, 2010.
Article in English | MEDLINE | ID: mdl-20409148

ABSTRACT

The ability to regulate wound contraction is critical for wound healing as well as for pathological contractures. Matrix metalloproteinases (MMPs) have been demonstrated to be obligatory for normal wound healing. This study examined the effect that the broad-spectrum MMP inhibitor BB-94 has when applied topically to full-thickness skin excisional wounds in rats and its ability to inhibit the promotion of myofibroblast formation and function by the latent transforming-growth factor-beta1 (TGF-beta1). BB-94 delayed wound contraction, as well as all other associated aspects of wound healing examined, including myofibroblast formation, stromal cell proliferation, blood vessel formation, and epithelial wound coverage. Interestingly, BB-94 dramatically increased the level of latent and active MMP-9. The increased levels of active MMP-9 may eventually overcome the ability of BB-94 to inhibit this MMP and may explain why wound contraction and other associated events of wound healing were only delayed and not completely inhibited. BB-94 was also found to inhibit the ability of latent TGF-beta1 to promote the formation and function of myofibroblasts. These results suggest that BB-94 could delay wound closure through a twofold mechanism; by blocking keratinocyte migration and thereby blocking the necessary keratinocyte-fibroblast interactions needed for myofibroblast formation and by inhibiting the activation of latent TGF-beta1.


Subject(s)
Phenylalanine/analogs & derivatives , Protease Inhibitors/pharmacology , Skin/cytology , Thiophenes/pharmacology , Wound Healing/drug effects , Wound Healing/physiology , Animals , Cell Proliferation/drug effects , Fibroblasts/drug effects , Matrix Metalloproteinase 9/drug effects , Neovascularization, Physiologic/drug effects , Phenylalanine/pharmacology , Rats , Stromal Cells/drug effects , Stromal Cells/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors
6.
Wound Repair Regen ; 14(3): 313-20, 2006.
Article in English | MEDLINE | ID: mdl-16808810

ABSTRACT

During wound healing and fibrocontractive diseases fibroblasts acquire a smooth muscle cell-like phenotype by differentiating into contractile force generating myofibroblasts. We examined whether regulation of myofibroblast contraction in granulation tissue is dominated by Ca2+-induced phosphorylation of myosin light chain kinase or by Rho/Rho kinase (ROCK)-mediated inhibition of myosin light chain phosphatase, similar to that of cultured myofibroblasts. Strips of granulation tissue obtained from rat granuloma pouches were stimulated with endothelin-1 (ET-1), serotonin, and angiotensin-II and isometric force generation was measured. We here investigated ET-1 in depth, because it was the only agonist that produced a long-lasting and strong response. The ROCK inhibitor Y27632 completely inhibited ET-1-promoted contraction and the phosphatase inhibitor calyculin elicited contraction in the absence of any other agonists, suggesting that activation of the Rho/ROCK/myosn light chain phosphatase pathway is critical in regulating in vivo myofibroblast contraction. Membrane depolarization with K+ also stimulated a long-lasting contraction of granulation tissue; however, the amount of force generated was significantly less compared to ET-1. Moreover, K+-induced contraction was inhibited by Y27632. These results are consistent with inhibition of myosin light chain phosphatase by the Rho/ROCK signaling pathway, which would account for the long-duration contraction of myofibroblasts necessary for wound closure.


Subject(s)
Fibroblasts/physiology , Granulation Tissue/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Isometric Contraction , Muscle, Smooth/cytology , Myosin-Light-Chain Phosphatase/metabolism , Protein Serine-Threonine Kinases/metabolism , Wound Healing/physiology , Amides/pharmacology , Animals , Endothelin-1/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Granulation Tissue/enzymology , Granulation Tissue/pathology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Isometric Contraction/drug effects , Male , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , rho-Associated Kinases
7.
Invest Ophthalmol Vis Sci ; 47(6): 2693-700, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16723488

ABSTRACT

PURPOSE: Vascular smooth muscle (SM) cells and pericytes are essential for normal vascular development. SM alpha-actin null mice were used to determine whether vascular SM and pericyte contractile functions, and not merely their presence, are necessary for vascular development, normal blood-retina barrier (BRB) permeability, and retinal function. METHODS: Age-matched SM alpha-actin null and wild-type mice were analyzed. Retinal structure, vascular pattern, and SM cell and pericyte distribution were analyzed histologically. Retinal vascular permeability (RVP) was measured with the Evans blue dye method. Electroretinography (ERG) was performed to evaluate retinal function. RESULTS: Deletion of SM alpha-actin did not result in any alterations in retinal morphology, vascular pattern, or SM cell and pericyte ensheathing of vessels in SM alpha-actin null mice. A significant increase in RVP was observed in SM alpha-actin null mice at both postnatal day (P)50 and P75 (P<0.05 and P<0.001, respectively). ERG analysis demonstrated a significant reduction in both rod and cone function in SM alpha-actin null mice at P22, P45, and P75 (P<0.01 at all ages). CONCLUSIONS: These results demonstrate that SM alpha-actin in SM cells and pericytes is not necessary for the formation of a normal retinal vascular pattern; however, SM alpha-actin is necessary for SM cells and pericytes to interact with endothelial cells to form a fully functional BRB. These results are important in understanding the role of contractile gene expression in the maintenance and function of the BRB and may provide a model for studying pathologic conditions, such as diabetes, that alter the function of this barrier.


Subject(s)
Actins/physiology , Blood-Retinal Barrier/physiology , Capillary Permeability/physiology , Gene Silencing/physiology , Muscle, Smooth, Vascular/physiology , Retina/physiology , Retinal Vessels/metabolism , Albumins/metabolism , Animals , Blotting, Western , Electroretinography , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Pericytes/physiology
8.
Am J Pathol ; 166(5): 1343-51, 2005 May.
Article in English | MEDLINE | ID: mdl-15855636

ABSTRACT

Myofibroblasts are specialized contractile fibroblasts that are critical in wound closure and tissue contracture. Generation of contractile force is correlated with the expression of alpha-smooth muscle actin (alpha-SMA); however, little is known regarding molecular mechanisms that control activation of alpha-SMA in myofibroblasts in granulation tissue. The aims of the present studies were to identify sufficient promoter regions required for alpha-SMA expression in myofibroblasts in vivo and to determine whether activation of alpha-SMA expression in myofibroblasts in vivo is dependent on an intronic CArG [CC(A/T)6GG] and a transforming growth factor-beta1 control element (TCE) that are required for alpha-SMA expression in smooth muscle cells. A Lac Z transgene construct from -2600 through the first intron was expressed in myofibroblasts within granulation tissue of cutaneous wounds in a pattern that closely mimicked endogenous alpha-SMA expression. Mutation of either the intronic CArG element or the TCE completely inhibited transgene expression in myofibroblasts in granulation tissue and responsiveness to transforming growth factor-beta1 in cultured transgenic fibroblasts. These same elements were also critical in regulating alpha-SMA expression during skeletal muscle repair but not during skeletal muscle development. Taken together, these results provide the first in vivo evidence for the importance of the intronic CArG and TCE cis-elements in the regulation of alpha-SMA expression in myofibroblasts in granulation tissue.


Subject(s)
Actins/genetics , Actins/metabolism , Fibroblasts/metabolism , Genes, Regulator/physiology , Granulation Tissue/metabolism , Introns/physiology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Transforming Growth Factor beta/genetics , Animals , Gene Expression Regulation , Genes, Reporter , Granulation Tissue/cytology , Male , Mice , Mice, Transgenic , Mutation/physiology , Promoter Regions, Genetic/physiology , Rats , Serum Response Factor/metabolism , Transforming Growth Factor beta1
9.
Exp Cell Res ; 299(2): 465-75, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15350544

ABSTRACT

In this study, we examined the impact of matrix metalloproteinases (MMP) on epithelialization, granulation tissue development, wound contraction, and alpha-smooth muscle actin (ASMA) expression during cutaneous wound repair through systemic administration of the synthetic broad-spectrum MMP inhibitor GM 6001 (N-[(2R)-2-(hydroxamidocarbonylmethyl)-4-methylpentanoyl]-L-tryptophan methylamide). Four full-thickness excisional wounds (50 mm2) on the back of 22 young female Sprague-Dawley rats, 12 treated with GM 6001 100 mg/kg and 10 with vehicle, were allowed to heal by secondary intention. GM 6001-treated wounds were minimally resurfaced with neoepithelium, despite unaltered keratinocyte proliferation in wound edges, whereas control wounds were completely covered with 3-7 cell layers of parakeratinized epithelium on post-wounding day 7. Hydroxyproline concentration, a marker of collagen, and cell proliferation in granulation tissue did not differ significantly between GM 6001-treated and control groups. Impaired wound contraction (P < 0.01) was associated with a dramatic reduction of ASMA-positive myofibroblasts in granulation tissue of GM 6001 wounds. This was not due to GM6001 blocking transforming growth factor-beta1 (TGF-beta1)-induced myofibroblast differentiation since GM 6001 did not inhibit TGF-beta1-induced ASMA expression and force generation in cultured rat dermal fibroblasts. The profound impairment of skin repair by the nonselective MMP inhibitor GM 6001 suggests that keratinocyte resurfacing, wound contraction, and granulation tissue organization are highly MMP-dependent processes.


Subject(s)
Cell Movement , Dipeptides/pharmacology , Keratinocytes/physiology , Protease Inhibitors/pharmacology , Skin/cytology , Skin/drug effects , Wound Healing , Animals , Epithelial Cells/physiology , Female , Fibroblasts/physiology , Granulation Tissue , Keratinocytes/drug effects , Matrix Metalloproteinase Inhibitors , Muscle Cells , Muscle Contraction , Rats , Rats, Sprague-Dawley , Skin/injuries , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...