Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 40(7): 111192, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35977484

RESUMO

Fibroblasts differentiate into myofibroblasts by acquiring new contractile function. This is important for tissue repair, but it also contributes to organ fibrosis. Platelet-derived growth factor (PDGF) promotes tissue repair and fibrosis, but the relationship between PDGF and myofibroblasts is unclear. Using mice with lineage tracing linked to PDGF receptor α (PDGFRα) gene mutations, we examine cell fates during skin wound healing. Elevated PDGFRα signaling increases proliferation but unexpectedly delays the fibroblast-to-myofibroblast transition, suggesting that PDGFRα must be downregulated for myofibroblast differentiation. In contrast, deletion of PDGFRα decreases proliferation and myofibroblast differentiation by reducing serum response factor (SRF) nuclear localization. Consequences of SRF deletion resemble PDGFRα deletion, but deletion of two SRF coactivators, MRTFA and MRTFB, specifically eliminates myofibroblasts. Our findings suggest a scenario where PDGFRα signaling initially supports proliferation of fibroblast progenitors to expand their number during early wound healing but, later, PDGFRα downregulation facilitates fibroblast differentiation into myofibroblasts.


Assuntos
Miofibroblastos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Animais , Diferenciação Celular/fisiologia , Fibroblastos/metabolismo , Fibrose , Camundongos , Miofibroblastos/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Cicatrização
2.
J Extracell Vesicles ; 9(1): 1692417, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31807237

RESUMO

Tumour metastasis suppressor KAI1/CD82 inhibits tumour cell movement. As a transmembrane protein, tetraspanin CD82 bridges the interactions between membrane microdomains of lipid rafts and tetraspanin-enriched microdomains (TEMs). In this study, we found that CD82 and other tetraspanins contain cholesterol recognition/interaction amino-acid consensus (CRAC) sequences in their transmembrane domains and revealed that cholesterol binding of CD82 determines its interaction with lipid rafts but not with TEMs. Functionally, CD82 needs cholesterol binding to inhibit solitary migration, collective migration, invasion and infiltrative outgrowth of tumour cells. Importantly, CD82-cholesterol/-lipid raft interaction not only promotes extracellular release of lipid raft components such as cholesterol and gangliosides but also facilitates extracellular vesicle (EV)-mediated release of ezrin-radixin-moesin (ERM) protein Ezrin. Since ERM proteins link actin cytoskeleton to the plasma membrane, we show for the first time that cell movement can be regulated by EV-mediated releases, which disengage the plasma membrane from cytoskeleton and then impair cell movement. Our findings also conceptualize that interactions between membrane domains, in this case converge of lipid rafts and TEMs by CD82, can change cell movement. Moreover, CD82 coalescences with both lipid rafts and TEMs are essential for its inhibition of tumour cell movement and for its enhancement of EV release. Finally, our study underpins that tetraspanins as a superfamily of functionally versatile molecules are cholesterol-binding proteins. Abbreviations: Ab: antibody; CBM: cholesterol-binding motif; CCM: cholesterol consensus motif; CRAC/CARC: cholesterol recognition or interaction amino-acid consensus; CTxB: cholera toxin B subunit; ECM: extracellular matrix; ERM: ezrin, radixin and moesin; EV: extracellular vesicles; FBS: foetal bovine serum; mAb: monoclonal antibody; MST: microscale thermophoresis; pAb: polyclonal antibody; and TEM: tetraspanin-enriched microdomain.

3.
Int J Cancer ; 143(5): 1162-1175, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29577277

RESUMO

Renal cell carcinoma (RCC) is a common and devastating disease characterized by a hypoxic microenvironment, epithelial-mesenchymal transition and potent resistance to therapy evidencing the presence of cancer stem cells (CSCs). Various CSC markers have been studied in RCC, but overall there is limited data on their role and most markers studied have been relatively nonspecific. Doublecortin-like kinase 1 (DCLK1) is a validated CSC marker in the gastrointestinal tract and evidence for an equivalent role in other cancers is accumulating. We used bioinformatics, immunohistochemistry, flow cytometry, spheroid self-renewal and chemoresistance assays in combination with overexpression and siRNA-knockdown to study the stem cell-supportive role of DCLK1 alternative splice variants (DCLK1 ASVs) in RCC. To target tumor cells expressing DCLK1 ASVs directly, we developed a novel monoclonal antibody (CBT-15) and delivered it systemically to RCC tumor xenografts. DCLK1 ASVs were overexpressed, enriched together with CSC markers and predictive of overall and recurrence-free survival in RCC patients. In vitro, DCLK1 ASVs were able to directly stimulate essential molecular and functional characteristics of renal CSCs including expression of aldehyde dehydrogenase, self-renewal and resistance to FDA-approved receptor tyrosine kinase and mTOR inhibitors, while targeted downregulation of DCLK1 reversed these characteristics. Finally, targeting DCLK1 ASV-positive cells with the novel CBT-15 monoclonal antibody blocked RCC tumorigenesis in vivo. These findings establish DCLK1 as a CSC marker with implications for therapy, disease progression and survival in RCC and demonstrate the therapeutic value of DCLK1-targeted monoclonal antibodies against renal CSCs.


Assuntos
Processamento Alternativo , Carcinoma de Células Renais/patologia , Transformação Celular Neoplásica/patologia , Resistencia a Medicamentos Antineoplásicos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Renais/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Serina-Treonina Quinases/genética , Animais , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Quinases Semelhantes a Duplacortina , Transição Epitelial-Mesenquimal , Seguimentos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Masculino , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , RNA Interferente Pequeno/genética , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Matrix Biol ; 65: 59-74, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28797711

RESUMO

After skin injury fibroblasts migrate into the wound and transform into contractile, extracellular matrix-producing myofibroblasts to promote skin repair. Persistent activation of myofibroblasts can cause excessive fibrotic reactions, but the underlying mechanisms are not fully understood. We used SMA-GFP transgenic mice to study myofibroblast recruitment and activation in skin wounds. Myofibroblasts were initially recruited to wounds three days post injury, their number reached a maximum after seven days and subsequently declined. Expression profiling showed that 1749 genes were differentially expressed in sorted myofibroblasts from wounds seven days post injury. Most of these genes were linked with the extracellular region and cell periphery including genes encoding for extracellular matrix proteins. A unique panel of core matrisome and matrisome-associated genes was differentially expressed in myofibroblasts and several genes not yet known to be linked to myofibroblast-mediated wound healing were found (e.g. Col24a1, Podnl1, Bvcan, Tinagl1, Thbs3, Adamts16, Adamts19, Cxcl's, Ccl's). In addition, a complex network of G protein-coupled signaling events was regulated in myofibroblasts (e.g. Adcy1, Plbc4, Gnas). Hence, this first characterization of a myofibroblast-specific expression profile at the peak of in situ granulation tissue formation provides important insights into novel target genes that may control excessive ECM deposition during fibrotic reactions.


Assuntos
Actinas/genética , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Pele/lesões , Actinas/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Miofibroblastos/química , Miofibroblastos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Pele/citologia , Pele/metabolismo
5.
Cancer Res ; 76(14): 4090-9, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27287716

RESUMO

Doublecortin-like kinase 1 (DCLK1) is a gastrointestinal (GI) tuft cell kinase that has been investigated as a biomarker of cancer stem-like cells in colon and pancreatic cancers. However, its utility as a biomarker may be limited in principle by signal instability and dilution in heterogeneous tumors, where the proliferation of diverse tumor cell lineages obscures the direct measurement of DCLK1 activity. To address this issue, we explored the definition of a miRNA signature as a surrogate biomarker for DCLK1 in cancer stem-like cells. Utilizing RNA/miRNA-sequencing datasets from the Cancer Genome Atlas, we identified a surrogate 15-miRNA expression signature for DCLK1 activity across several GI cancers, including colon, pancreatic, and stomach cancers. Notably, Cox regression and Kaplan-Meier analysis demonstrated that this signature could predict the survival of patients with these cancers. Moreover, we identified patient subgroups that predicted the clinical utility of this DCLK1 surrogate biomarker. Our findings greatly strengthen the clinical significance for DCLK1 expression across GI cancers. Further, they provide an initial guidepost toward the development of improved prognostic biomarkers or companion biomarkers for DCLK1-targeted therapies to eradicate cancer stem-like cells in these malignancies. Cancer Res; 76(14); 4090-9. ©2016 AACR.


Assuntos
Neoplasias Gastrointestinais/mortalidade , Peptídeos e Proteínas de Sinalização Intracelular/análise , MicroRNAs/análise , Células-Tronco Neoplásicas/enzimologia , Proteínas Serina-Treonina Quinases/análise , Linhagem Celular Tumoral , Quinases Semelhantes a Duplacortina , Transição Epitelial-Mesenquimal , Neoplasias Gastrointestinais/patologia , Humanos
6.
Oncotarget ; 6(4): 2193-205, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25605241

RESUMO

Renal clear cell carcinoma (RCC) is the most common type of kidney cancer and the 8th most common cancer overall in the US. RCC survival rates drop precipitously with regional and distant spread and recent studies have demonstrated that RCC presents an epithelial-mesenchymal transition (EMT) phenotype linked to increased recurrence and decreased survival. EMT is a key characteristic of tumor stem cells (TSCs) along with chemo-resistance and radio-resistance, which are also phenotypic of RCC. Targeting these factors is key to increasing the survival of RCC patients. Doublecortin-like kinase 1 (DCLK1) marks TSCs in pancreatic and colorectal cancer and regulates EMT and stemness. Analysis of the Cancer Genome Atlas' RCC dataset revealed that DCLK1 is overexpressed and dysregulated on the mRNA and epigenetic level in more than 93% of RCC tumors relative to adjacent normal tissue. Immunohistochemistry using α-DCLK1 antibody confirmed overexpression and demonstrated a major increase in immunoreactivity in stage II-III tumors compared to normal kidney and stage I tumors. Small-interfering RNA (siRNA) mediated knockdown of DCLK1 resulted in decreased expression of EMT and pluripotency factors and significantly reduced invasion, migration, focal adhesion, drug-resistance, and clonogenic capacity. These findings suggest that DCLK1 is a novel, overexpressed factor in RCC progression that may be targeted to suppress EMT, metastasis, and stemness in early-stage and advanced RCC to increase patient survival. Moreover, the possibility that DCLK1 may mark a population of tumor stem-like cells in RCC should be further investigated in light of these findings.


Assuntos
Carcinoma de Células Renais/genética , Transição Epitelial-Mesenquimal/genética , Adesões Focais/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Renais/genética , Proteínas Serina-Treonina Quinases/genética , Western Blotting , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Metilação de DNA , Quinases Semelhantes a Duplacortina , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Invest Dermatol ; 134(6): 1725-1734, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24463424

RESUMO

Wound healing, angiogenesis, and hair follicle maintenance are often impaired in the skin of diabetic patients, but the pathogenesis has not been well understood. Here, we report that circulation levels of kallistatin, a member of the serine proteinase inhibitor (SERPIN) superfamily with antiangiogenic activities, were elevated in type 2 diabetic patients with diabetic vascular complications. To test the hypothesis that elevated kallistatin levels could contribute to a wound-healing deficiency via the inhibition of Wnt/ß-catenin signaling, we generated kallistatin-transgenic (KS-TG) mice. KS-TG mice had reduced cutaneous hair-follicle density, microvascular density, and panniculus adiposus layer thickness, as well as altered skin microvascular hemodynamics and delayed cutaneous wound healing. Using Wnt reporter mice, our results showed that Wnt/ß-catenin signaling is suppressed in the dermal endothelium and hair follicles in KS-TG mice. Lithium, a known activator of ß-catenin via inhibition of glycogen synthase kinase-3ß, reversed the inhibition of Wnt/ß-catenin signaling by kallistatin and rescued the wound-healing deficiency in KS-TG mice. These observations suggest that elevated circulating antiangiogenic serpins in diabetic patients may contribute to impaired wound healing through inhibition of Wnt/ß-catenin signaling. Activation of Wnt/ß-catenin signaling, at a level downstream of Wnt receptors, may ameliorate the wound-healing deficiency in diabetic patients.


Assuntos
Angiopatias Diabéticas/metabolismo , Regulação da Expressão Gênica , Serpinas/metabolismo , Via de Sinalização Wnt , Cicatrização , Adulto , Animais , Células Cultivadas , Diabetes Mellitus Tipo 2 , Feminino , Genes Reporter , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Folículo Piloso/metabolismo , Humanos , Lítio/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Microcirculação , Pessoa de Meia-Idade , Neovascularização Patológica , Serpinas/sangue , Serpinas/genética , Pele/irrigação sanguínea , Pele/metabolismo , Transcrição Gênica
8.
Adv Wound Care (New Rochelle) ; 2(4): 122-141, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-24527336

RESUMO

SIGNIFICANCE: Myofibroblasts are responsible for wound closure that occurs in healed acute wounds. However, their actions can result in disfiguring scar contractures, compromised organ function, and a tumor promoting stroma. Understanding the mechanisms regulating their contractile machinery, gene expression, and lifespan is essential to develop new therapies to control their function. RECENT ADVANCES: Mechanical stress and transforming growth factor beta-1 (TGF-ß1) regulate myofibroblast differentiation from mesenchymal progenitors. As these precursor cells differentiate, they assemble a contractile apparatus to generate the force used to contract wounds. The mechanisms by which mechanical stress promote expression of contractile genes through the TGF-ß1 and serum response factor pathways and offer therapeutic targets to limit myofibroblast function are being elucidated. CRITICAL ISSUES: Emerging evidence suggests that the integration of mechanical cues with intracellular signaling pathways is critical to myofibroblast function via its effects on gene expression, cellular contraction, and paracrine signaling with neighboring cells. In addition, while apoptosis is clearly one pathway that can limit myofibroblast lifespan, recent data suggest that pathogenic myofibroblasts can become senescent and adopt a more beneficial phenotype, or may revert to a quiescent state, thereby limiting their function. FUTURE DIRECTIONS: Given the important role that myofibroblasts play in pathologies as disparate as cutaneous scarring, organ fibrosis, and tumor progression, knowledge gained in the areas of intracellular signaling networks, mechanical signal transduction, extracellular matrix biology, and cell fate will support efforts to develop new therapies with a wide impact.

9.
Wound Repair Regen ; 21(1): 166-76, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23253249

RESUMO

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.


Assuntos
Actinas/metabolismo , Fibroblastos/patologia , Adesões Focais/patologia , Tecido de Granulação/patologia , Miofibroblastos/patologia , Cicatrização , Ferimentos e Lesões/patologia , Animais , Western Blotting , Diferenciação Celular , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
10.
Exp Cell Res ; 318(13): 1542-53, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22449415

RESUMO

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.


Assuntos
Fibroblastos/enzimologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Miofibroblastos/enzimologia , Actinas/química , Actinas/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Movimento Celular/genética , Movimento Celular/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Fator de Crescimento Insulin-Like I/farmacologia , Modelos Biológicos , Miofibroblastos/citologia , Miofibroblastos/fisiologia , Fenótipo , Multimerização Proteica , Interferência de RNA , Ratos , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Cicatrização/genética , Cicatrização/fisiologia
11.
J Invest Dermatol ; 131(12): 2378-85, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21776010

RESUMO

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.


Assuntos
Diferenciação Celular/genética , Fibroblastos/citologia , Miofibroblastos/citologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Proteínas do Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Adesões Focais/metabolismo , Miofibroblastos/metabolismo , Ratos
12.
Biol Reprod ; 85(1): 13-21, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21368298

RESUMO

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.


Assuntos
Actinas/metabolismo , Glândulas Mamárias Animais/fisiologia , Ejeção Láctea , Animais , Animais Recém-Nascidos , Insuficiência de Crescimento , Feminino , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Knockout , Ocitocina/fisiologia , Gravidez
13.
J Hand Surg Am ; 35(10): 1580-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20888494

RESUMO

PURPOSE: Previous studies suggest that Dupuytren's disease is caused by fibroblast and myofibroblast contractility within Dupuytren's nodules; however, the stimulus for cell contractility is unknown. Sphingosine-1-phosphate (S1P) is a serum-derived lysophospholipid mediator that enhances cell contractility by activating the S1P receptor, S1P(2). It is hypothesized that S1P stimulates Dupuytren's fibroblast contractility through S1P(2) activation of non-muscle myosin II (NMMII). This investigation examined the role of S1P and NMMII activation in Dupuytren's disease progression and suggests potential targets for treatment. METHODS: We enmeshed Dupuytren's fibroblasts into fibroblast-populated collagen lattices (FPCLs) and assayed S1P-stimulated FPCL contraction in the presence of the S1P(2) receptor inhibitor JTE-013, the Rho kinase inhibitor Y-27632, the myosin light chain kinase inhibitor ML-7, and the NMMII inhibitor blebbistatin. Tissues from Dupuytren's fascia (n = 10) and normal palmar fascia (n = 10) were immunostained for NMMIIA and NMMIIB. RESULTS: Sphingosine-1-phosphate stimulated FPCL contraction in a dose-dependent manner. Inhibition of S1P(2) and NMMII prevented S1P-stimulated FPCL contraction. Rho kinase and myosin light chain kinase inhibited both S1P and control FPCL contraction. Dupuytren's nodule fibroblasts robustly expressed NMMIIA and NMMIIB, compared with quiescent-appearing cords and normal palmar fascia. CONCLUSIONS: Sphingosine-1-phosphate promotes Dupuytren's fibroblast contractility through S1P(2), which stimulates activation of NMMII. NMMII isoforms are ubiquitously expressed throughout Dupuytren's nodules, which suggests that nodule fibroblasts are primed to respond to S1P stimulation to cause contracture formation. S1P-promoted activation of NMMII may be a target for disease treatment.


Assuntos
Contratura de Dupuytren/metabolismo , Fibroblastos/química , Lisofosfolipídeos/metabolismo , Músculo Liso/química , Miosina Tipo II/biossíntese , Esfingosina/análogos & derivados , Linhagem Celular , Colágeno , Progressão da Doença , Fibroblastos/fisiologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Técnicas Imunoenzimáticas , Contração Muscular , Esfingosina/metabolismo
14.
Wound Repair Regen ; 18(2): 223-34, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20409148

RESUMO

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.


Assuntos
Fenilalanina/análogos & derivados , Inibidores de Proteases/farmacologia , Pele/citologia , Tiofenos/farmacologia , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fenilalanina/farmacologia , Ratos , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores
15.
Am J Physiol Cell Physiol ; 298(1): C191-201, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19846754

RESUMO

During platelet-derived growth factor (PDGF)-BB-mediated recruitment to neovascular sprouts, vascular smooth muscle cells (VSMCs) dedifferentiate from a contractile to a migratory phenotype. This involves the downregulation of contractile markers such as smooth muscle (SM) alpha-actin and the upregulation of promigration genes such as matrix metalloproteinase (MMP)-2. The regulation of MMP-2 in response to PDGF-BB is complex and involves both stimulatory and inhibitory signaling pathways, resulting in a significant delay in upregulation. Here, we provide evidence that the delay in MMP-2 upregulation may be due to the autocrine expression and activation of transforming growth factor (TGF)-beta, which is known to promote the contractile phenotype in VSMCs. Whereas PDGF-BB could induce the loss of stress fibers and focal adhesions, TGF-beta was able to block or reverse this transition to a noncontractile state. TGF-beta did not, however, suppress early signaling events stimulated by PDGF-BB. Over time, though PDGF-BB induced increased TGF-beta1 levels, it suppressed TGF-beta2 and TGF-beta3 expression, leading to a net decrease in the total TGF-beta pool, resulting in the upregulation of MMP-2. Together, these findings indicate that MMP-2 expression is suppressed by a threshold level of active TGF-beta, which in turn promotes a contractile VSMC phenotype that prevents the upregulation of MMP-2.


Assuntos
Metaloproteinase 2 da Matriz/genética , Músculo Liso Vascular/enzimologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima/efeitos dos fármacos , Actinas/genética , Animais , Becaplermina , Primers do DNA , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Genes fos , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Endogâmicos WKY , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-2/genética
16.
Wound Repair Regen ; 14(3): 313-20, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16808810

RESUMO

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.


Assuntos
Fibroblastos/fisiologia , Tecido de Granulação/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Contração Isométrica , Músculo Liso/citologia , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Cicatrização/fisiologia , Amidas/farmacologia , Animais , Endotelina-1/farmacologia , Inibidores Enzimáticos/farmacologia , Fibroblastos/enzimologia , Tecido de Granulação/enzimologia , Tecido de Granulação/patologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Contração Isométrica/efeitos dos fármacos , Masculino , Fosfatase de Miosina-de-Cadeia-Leve/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Quinases Associadas a rho
17.
Invest Ophthalmol Vis Sci ; 47(6): 2693-700, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16723488

RESUMO

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.


Assuntos
Actinas/fisiologia , Barreira Hematorretiniana/fisiologia , Permeabilidade Capilar/fisiologia , Inativação Gênica/fisiologia , Músculo Liso Vascular/fisiologia , Retina/fisiologia , Vasos Retinianos/metabolismo , Albuminas/metabolismo , Animais , Western Blotting , Eletrorretinografia , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pericitos/fisiologia
18.
J Invest Dermatol ; 126(3): 561-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16397521

RESUMO

IL-6 deficient (IL-6KO) mice display significantly delayed cutaneous wound closure. Myofibroblasts are the primary mediators of wound closure, and alpha-smooth muscle actin (alpha-SMA) is a marker of fibroblast differentiation to the myofibroblast phenotype. Wounds from IL-6KO, and wild-type mice were collected up to 6 days following wounding. Expression of alpha-SMA mRNA was found to be increased in wounds of IL-6KO mice up to 48 hours post wounding, but decreased below wild-type levels by 72 hours. Recombinant IL-6 treatment of IL-6KO dermal fibroblasts showed an induction of alpha-SMA mRNA and protein peaking at 1 ng/ml cytokine, but declining at higher concentrations. Actinomycin-D treatment of fibroblast cultures indicated that recombinant mouse IL-6 (rmIL-6) induction of alpha-SMA mRNA appeared to be primarily transcriptionally regulated, and extracellular signal-regulated kinase 1/2 kinase, but not signal transducers and activators of transcription 3 was readily phosphorylated in rmIL-6 treated IL-6KO fibroblasts. A dose-response increase in the mRNA expression of the IL-6R signaling inhibitor protein suppressors of cytokine signaling (SOCS) 3 was also noted in rmIL-6-treated IL-6KO fibroblasts. These data indicate that alpha-SMA expression is dysregulated in IL-6KO mice. The expression of alpha-SMA induced by rmIL-6 in fibroblasts from IL-6KO mice appears to be transcriptionally modulated, dependent on JAK1 kinase, and possibly downregulated as a result of increased SOCS3 expression.


Assuntos
Actinas/genética , Regulação da Expressão Gênica , Interleucina-6/fisiologia , Pele/metabolismo , Cicatrização , Animais , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Fibroblastos/metabolismo , Janus Quinase 1 , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Tirosina Quinases/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Ativação Transcricional
19.
Am J Pathol ; 166(5): 1343-51, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15855636

RESUMO

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.


Assuntos
Actinas/genética , Actinas/metabolismo , Fibroblastos/metabolismo , Genes Reguladores/fisiologia , Tecido de Granulação/metabolismo , Íntrons/fisiologia , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Crescimento Transformador beta/genética , Animais , Regulação da Expressão Gênica , Genes Reporter , Tecido de Granulação/citologia , Masculino , Camundongos , Camundongos Transgênicos , Mutação/fisiologia , Regiões Promotoras Genéticas/fisiologia , Ratos , Fator de Resposta Sérica/metabolismo , Fator de Crescimento Transformador beta1
20.
Exp Cell Res ; 299(2): 465-75, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15350544

RESUMO

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.


Assuntos
Movimento Celular , Dipeptídeos/farmacologia , Queratinócitos/fisiologia , Inibidores de Proteases/farmacologia , Pele/citologia , Pele/efeitos dos fármacos , Cicatrização , Animais , Células Epiteliais/fisiologia , Feminino , Fibroblastos/fisiologia , Tecido de Granulação , Queratinócitos/efeitos dos fármacos , Inibidores de Metaloproteinases de Matriz , Células Musculares , Contração Muscular , Ratos , Ratos Sprague-Dawley , Pele/lesões , Fator de Crescimento Transformador beta/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...