Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Pathol ; 190(1): 206-221, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31610176

RESUMEN

Tumor stroma resembles a fibrotic microenvironment, being characterized by the presence of myofibroblast-like cancer-associated fibroblasts (CAFs). In wild-type mice injected with melanoma cells, we show that the stem cell transcription factor Sox2 is expressed by tumor cells and induced in CAFs derived from synthetic fibroblasts. These fibroblasts were labeled postnatally with green fluorescent protein using mice expressing a tamoxifen-dependent Cre recombinase under the control of a fibroblast-specific promoter/enhancer. Conversely, fibroblast activation was impaired in mice with a fibroblast-specific deletion of cellular communication network 2 (Ccn2), associated with reduced expression of α-smooth muscle actin and Sox2. Multipotent Sox2-expressing skin-derived precursor (SKP) spheroids were cultured from murine back skin. Using lineage tracing and flow cytometry, approximately 40% of SKPs were found to be derived from type I collagen-lineage cells and acquired multipotency in culture. Inhibition of mechanotransduction pathways prevented myofibroblast differentiation of SKPs and expression of Ccn2. In SKPs deleted for Ccn2, differentiation into a myofibroblast, but not an adipocyte or neuronal phenotype, was also impaired. In human melanoma, CCN2 expression was associated with a profibrotic integrin alpha (ITGA) 11-expressing subset of CAFs that negatively associated with survival. These results suggest that synthetic dermal fibroblasts are plastic, and that CCN2 is required for the differentiation of dermal progenitor cells into a myofibroblast/CAF phenotype and is, therefore, a therapeutic target in melanoma.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Fibroblastos/patología , Fibrosis/patología , Melanoma Experimental/patología , Piel/patología , Células Madre/patología , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Diferenciación Celular , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibroblastos/metabolismo , Fibrosis/metabolismo , Humanos , Mecanotransducción Celular , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Noqueados , Pronóstico , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Piel/metabolismo , Células Madre/metabolismo , Tasa de Supervivencia , Microambiente Tumoral
2.
Cancer Res Commun ; 4(2): 556-570, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-38363129

RESUMEN

Melanoma is the leading cause of skin cancer-related death. As prognosis of patients with melanoma remains problematic, identification of new therapeutic targets remains essential. Matricellular proteins are nonstructural extracellular matrix proteins. They are secreted into the tumor microenvironment to coordinate behavior among different cell types, yet their contribution to melanoma is underinvestigated. Examples of matricellular proteins include those comprising the CCN family. The CCN family member, CCN1, is highly proangiogenic. Herein, we show that, in human patients with melanoma, although found in several tumor cell types, CCN1 is highly expressed by a subset of cancer-associated fibroblasts (CAF) in patients with melanoma and this expression correlates positively with expression of proangiogenic genes and progressive disease/resistance to anti-PD1 checkpoint inhibitors. Consistent with these observations, in a syngeneic C57BL6 mouse model of melanoma, loss of CCN1 expression from Col1A2-Cre-, herein identified as "universal," fibroblasts, impaired metastasis of subcutaneously injected B16F10 tumor cells to lung, concomitant with disrupted neovascularization and collagen organization. Disruption of the extracellular matrix in the loss of CCN1 was validated using a novel artificial intelligence-based image analysis platform that revealed significantly decreased phenotypic fibrosis and composite morphometric collagen scores. As drug resistance is linked to matrix deposition and neoangiogenesis, these data suggest that CCN1, due to its multifaceted role, may represent a novel therapeutic target for drug-resistant melanoma. Our data further emphasize the essential role that cancer-associated, (universal) Col1A2-Cre-fibroblasts and extracellular matrix remodeling play in coordinating behavior among different cell types within the tumor microenvironment. SIGNIFICANCE: In human patients, the expression of proangiogenic matricellular protein CCN1 in CAFs correlates positively with expression of stroma and angiogenic markers and progressive disease/resistance to checkpoint inhibitor therapy. In an animal model, loss of CCN1 from CAFs impaired metastasis of melanoma cells, neovascularization, and collagen deposition, emphasizing that CAFs coordinate cellular behavior in a tumor microenvironment and that CCN1 may be a novel target.


Asunto(s)
Fibroblastos Asociados al Cáncer , Melanoma , Animales , Humanos , Ratones , Inteligencia Artificial , Fibroblastos Asociados al Cáncer/metabolismo , Colágeno , Proteína 61 Rica en Cisteína/genética , Melanoma/genética , Neovascularización Patológica/genética , Microambiente Tumoral/genética
4.
Matrix Biol Plus ; 3: 100009, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33543008

RESUMEN

The microenvironment contributes to the excessive connective tissue deposition that characterizes fibrosis. Members of the CCN family of matricellular proteins are secreted by fibroblasts into the fibrotic microenvironment; however, the role of endogenous CCN1 in skin fibrosis is unknown. Mice harboring a fibroblast-specific deletion for CCN1 were used to assess if CCN1 contributes to dermal homeostasis, wound healing, and skin fibrosis. Mice with a fibroblast-specific CCN1 deletion showed progressive skin thinning and reduced accumulation of type I collagen; however, the overall mechanical property of skin (Young's modulus) was not significantly reduced. Real time-polymerase chain reaction analysis revealed that CCN1-deficient skin displayed reduced expression of mRNAs encoding enzymes that promote collagen stability (including prolyl-4-hydroxylase and PLOD2), although expression of COL1A1 mRNA was unaltered. CCN1-deficent skin showed reduced hydroxyproline levels. Electron microscopy revealed that collagen fibers were disorganized in CCN1-deficient skin. CCN1-deficient mice were resistant to bleomycin-induced skin fibrosis, as visualized by reduced collagen accumulation and skin thickness suggesting that deposition/accumulation of collagen is impaired in the absence of CCN1. Conversely, CCN1-deficient mice showed unaltered wound closure kinetics, suggesting de novo collagen production in response to injury did not require CCN1. In response to either wounding or bleomycin, induction of α-smooth muscle actin-positive myofibroblasts was unaffected by loss of CCN1. CCN1 protein was overexpressed by dermal fibroblasts isolated from lesional (i.e., fibrotic) areas of patients with early onset diffuse scleroderma. Thus, CCN1 expression by fibroblasts, being essential for skin fibrosis, is a viable anti-fibrotic target.

5.
J Cell Commun Signal ; 12(1): 113-118, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29110248

RESUMEN

Melanoma metastasis is fatal. Melanoma cells are often characterized by an activated extracellular signal-regulated kinase (ERK) pathway downstream of mutations in BRAF. Therapies targeting these BRAF mutations are useful for a while; however, patients ultimately develop resistance to these therapies. Recent evidence suggests that this resistance occurs when tumor cells leave their microenvironment and migrate on a stiff, activated tumor stroma; that is, this resistance is linked to the presence of an extracellular matrix reminiscent of a fibrotic micronvironment. These data suggest that agents targeting fibrosis might be used to treat melanoma. We therefore discuss what is known about the tumor stroma in melanoma. An emergent target, CCN2 (CTGF), that is required for fibrosis, may also be a good target for drug-resistant melanoma. Intriguingly, anti-CCN2 antibodies are currently under clinical development.

6.
Matrix Biol ; 74: 52-61, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29885461

RESUMEN

Metastatic melanoma is highly fatal. Within the tumor microenvironment, the role of cancer-associated fibroblasts (CAFs) in melanoma metastasis and progression is relatively understudied. The matricellular protein CCN2 (formerly termed connective tissue growth factor, CTGF) is overexpressed, in a fashion independent of BRAF mutational status, by CAFs in melanoma. Herein, we find, in human melanoma patients, that CCN2 expression negatively correlates with survival and positively correlates with expression of neovascularization markers. To assess the role of CAFs in melanoma progression, we used C57BL/6 mice expressing a tamoxifen-dependent cre recombinase expressed under the control of a fibroblast-specific promoter/enhancer (COL1A2) to delete CCN2 postnatally in fibroblasts. Mice deleted or not for CCN2 in fibroblasts were injected subcutaneously with B16-F10 melanoma cells. Loss of CCN2 in CAFs resulted in reduced CAF activation, as detected by staining with anti-α-smooth muscle actin antibodies, and reduced tumor-induced neovascularization, as detected by micro-computed tomography (micro-CT) and staining with anti-CD31 antibodies. CCN2-deficient B16(F10) cells were defective in a tubule formation/vasculogenic mimicry assay in vitro. Mice deleted for CCN2 in CAFs also showed impaired vasculogenic mimicry of subcutaneously-injected B16-F10 cells in vivo. Our results provide new insights into the cross-talk among different cell types in the tumor microenvironment and suggest CAFs play a heretofore unappreciated role by being essential for tumor neovascularization via the production of CCN2. Our data are consistent with the hypothesis that activated CAFs are essential for melanoma metastasis and that, due to its role in this process, CCN2 is a therapeutic target for melanoma.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Melanoma/irrigación sanguínea , Neovascularización Patológica/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Melanoma/diagnóstico por imagen , Melanoma/genética , Melanoma/metabolismo , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/genética , Pronóstico , Transducción de Señal , Análisis de Supervivencia , Microambiente Tumoral , Regulación hacia Arriba , Microtomografía por Rayos X
7.
Methods Mol Biol ; 1489: 423-429, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27734394

RESUMEN

CCN2 is a profibrotic matricellular protein. CCN2 directly promotes cell adhesion and indirectly promotes fibrosis by activating adhesive signaling in response to growth factors, cytokines, and extracellular matrix. The following protocols will allow the direct assessment of other CCN family members in these processes.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Animales , Adhesión Celular/genética , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibrosis/patología , Técnicas de Inactivación de Genes , Ratones , Ratones Transgénicos , Piel/metabolismo , Piel/patología
8.
PLoS One ; 10(4): e0123689, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25927238

RESUMEN

Transforming growth factor (TGF)ß acts on fibroblasts to promote the production and remodeling of extracellular matrix (ECM). In adult humans, excessive action of TGFß is associated with fibrotic disease and fibroproliferative conditions, including gingival hyperplasia. Understanding how the TGFß1 signals in fibroblasts is therefore likely to result in valuable insights into the fundamental mechanisms underlying fibroproliferative disorders. Previously, we used the TAK1 inhibitor (5Z)-7-Oxozeaenol to show that, in dermal fibroblasts, the non-canonical TAK1 pathway mediates the ability of TGFß1 to induce genes promoting tissue remodeling and repair. However, the extent to which TAK1 mediates fibroproliferative responses in fibroblasts in response to TGFß1 remains unclear. Herein, we show that, in gingival fibroblasts, (5Z)-7-Oxozeaenol blocks the ability of TGFß1 to induce expression of the pro-fibrotic mediator CCN2 (connective tissue growth factor, CTGF) and type I collagen protein. Moreover, genome-wide expression profiling revealed that, in gingival fibroblasts, (5Z)-7-Oxozeaenol reduces the ability of TGFß1 to induce mRNA expression of essentially all TGFß1-responsive genes (139/147), including those involved with a hyperproliferative response. Results from microarray analysis were confirmed using real time polymerase chain reaction analysis and a functional cell proliferation assay. Our results are consistent with the hypothesis that TAK1 inhibitors might be useful in treating fibroproliferative disorders, including that in the oral cavity.


Asunto(s)
Fibroblastos/metabolismo , Encía/metabolismo , Lactonas/farmacología , Resorcinoles/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Células Cultivadas , Colágeno Tipo I/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Femenino , Fibroblastos/citología , Regulación de la Expresión Génica/efectos de los fármacos , Encía/citología , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/biosíntesis , Masculino
9.
Matrix Biol ; 43: 35-41, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25644104

RESUMEN

Elevated adhesive signaling promotes fibrosis. Protein phosphatase and tensin homologue (PTEN) dephosphorylates focal adhesion kinase and suppresses the activation of Akt and hence suppresses adhesive signaling. Loss of PTEN expression is associated with lung fibrosis, but whether PTEN expression by type I collagen-expressing cells controls lung fibrosis is unclear. Here, we use mice expressing tamoxifen-dependent cre recombinase expressed under the control of a COL1A2 promoter/enhancer and mice harboring floxed-PTEN and/or floxed-CCN2 alleles to assess whether loss of PTEN expression by type I collagen producing cells results in lung fibrosis in a CCN2-dependent fashion. In vivo, loss of PTEN expression resulted in the overexpression of both collagen type I and the pro-adhesive matricellular protein connective tissue growth factor (CTGF/CCN2). However, α-smooth muscle actin expression was unaffected. Loss of CCN2 expression by lung fibroblasts rescues this phenotype; i.e.., mice deficient in both PTEN and CCN2 in collagen type I-expressing cells do not develop significant collagen deposition in the lung. PTEN expression by collagen type I-expressing cells controls collagen deposition; therapeutic strategies blocking CCN2 may be of benefit in blocking excessive collagen deposition in fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibroblastos/metabolismo , Fosfohidrolasa PTEN/genética , Fibrosis Pulmonar/patología , Animales , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibroblastos/patología , Regulación de la Expresión Génica , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Ratones , Fosfohidrolasa PTEN/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo
10.
J Invest Dermatol ; 135(11): 2805-2813, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26168233

RESUMEN

Metastatic melanoma has an extremely poor prognosis with few durable remissions. The secreted matricellular protein connective tissue growth factor (CCN2) is overexpressed in cancers including melanoma and may represent a viable therapeutic target. However, the mechanism underlying the contribution of CCN2 to melanoma progression is unclear. Herein, we use the highly metastatic murine melanoma cell line B16(F10) and syngeneic mice, in which CCN2 expression is knocked out in fibroblasts, to demonstrate that loss of CCN2, either in melanoma cells or in the niche, impedes the ability of melanoma cells to invade. Specifically, loss of CCN2 in melanoma cells diminished their ability to invade through collagen in vitro, and loss of fibroblast-derived CCN2 decreased spontaneous metastases of melanoma cells from the skin to the lungs in vivo. Proliferation and tumor growth were not affected by loss of CCN2. CCN2-deficient B16(F10) cells showed reduced expression of the matricellular protein periostin; addition of recombinant periostin rescued the in vitro invasion defect of these cells. Immunohistochemical analysis of CCN2-deficient mice confirmed loss of periostin expression in the absence of CCN2. CCN2 and periostin mRNA levels are positively correlated with each other and with the stromal composition of human melanoma lesions but not BRAF mutations. Thus, CCN2 promotes invasion and metastasis via periostin and should be further evaluated as a possible therapeutic target for BRAF inhibitor-resistant melanoma.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Regulación Neoplásica de la Expresión Génica , Metástasis de la Neoplasia/genética , Neoplasias Cutáneas/genética , Animales , Biopsia con Aguja , Línea Celular Tumoral , Progresión de la Enfermedad , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Neoplasias Cutáneas/patología , Células del Estroma/patología , Regulación hacia Arriba
11.
J Cell Commun Signal ; 8(1): 65-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24193793

RESUMEN

Melanoma is becoming increasingly common in recent years and has a very high mortality rate, owing largely to its highly metastatic nature. It tends to metastasize early in the course of the disease, and after metastasis is resistant to most current therapies. Preclinical data has indicated that heparin administered to patients as an antithrombotic treatment also has anti-metastatic properties. Heparin has been shown to interfere with the binding of the integrin Very Late Antigen 4 (VLA-4) to its ligand Vascular Cell Adhesion Molecule 1 (VCAM-1) and in a recent paper the laboratory of Bendas (Thrombosis and Haemostasis, Prepublished online) demonstrated that CCN1 binds to VLA-4 and interference in this binding by heparin results in reduced strength of the VLA-4/VCAM-1 binding. This indicates that CCN1 might represent a good target for reducing the metastasis, and thus mortality, of melanoma.

12.
J Cell Commun Signal ; 8(2): 171-2, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24458847

RESUMEN

Cell-mediated activation of latent TGF-ß1 is intimately involved with tissue repair and fibrosis in all organs. Previously, it was shown that the integrin ß1 subunit was required for activation of latent TGF-ß1 and skin fibrosis. A recent study by Henderson and colleagues (Nature Medicine 19,1617-1624, 2013) used three different in vivo models of fibrosis to show that integrin αv subunit was required for fibrogenesis. Through a process of elimination, the authors conclude that in vivo, the little-studied αvß1 could be the major integrin responsible for TGF-ß activation by myofibroblasts. Thus targeting this integrin might be a useful therapy for fibrosis.

13.
J Invest Dermatol ; 133(6): 1646-54, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23340735

RESUMEN

Dermal connective tissue is a supportive structure required for skin's barrier function; dysregulated dermal homeostasis results in chronic wounds and fibrotic diseases. The multifunctional cytokine transforming growth factor (TGF) ß promotes connective tissue deposition, repair, and fibrosis. TGF-ß acts through well-defined canonical pathways; however, the non-canonical pathways through which TGF-ß selectively promotes connective tissue deposition are unclear. In dermal fibroblasts, we show that inhibition of the non-canonical TGF-ß-activated kinase 1 (TAK1) selectively reduced the ability of TGF-ß to induce expression of a cohort of wound healing genes, such as collagens, CCN2, TGF-ß1, and IL-6. Fibroblast-specific TAK1-knockout mice showed impaired cutaneous tissue repair and decreased collagen deposition, α-smooth muscle actin and CCN2 expression, proliferating cell nuclear antigen staining, and c-Jun N-terminal kinase and p38, but not Smad3, phosphorylation. TAK1-deficient fibroblasts showed reduced cell proliferation, migration, cell attachment/spreading, and contraction of a floating collagen gel matrix. TAK1-deficient mice also showed progressively reduced skin thickness and collagen deposition. Thus, TAK1 is essential for connective tissue deposition in the dermis.


Asunto(s)
Dermis/citología , Dermis/fisiología , Homeostasis/fisiología , Quinasas Quinasa Quinasa PAM/fisiología , Cicatrización de Heridas/fisiología , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Fibroblastos/citología , Fibroblastos/fisiología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Noqueados , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Zearalenona/análogos & derivados , Zearalenona/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA