Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
iScience ; 27(6): 109864, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38770136

RESUMEN

Hippo was first identified in a genetic screen as a protein that suppressed proliferation and cell growth. Subsequently, it was shown that hippo acted in a so-called canonical cascade to suppress Yorkie, the Drosophila equivalent of Yes-activated protein (YAP), a mechanosensitive transcriptional cofactor that enhances the activity of the TEAD family of transcription factors. YAP promotes fibrosis, activation of cancer-associated fibroblasts, angiogenesis and cancer cell invasion. YAP activates the expression of the matricellular proteins CCN1 (cyr61) and CCN2 (ctgf), themselves mediators of fibrogenesis and oncogenesis, and coordination of matrix deposition and angiogenesis. This review discusses how therapeutically targeting YAP through YAP inhibitors verteporfin and celastrol and its downstream mediators CCN1 and CCN2 might be useful in treating melanoma.

2.
Expert Opin Investig Drugs ; 33(3): 279-285, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38393748

RESUMEN

INTRODUCTION: The potent profibrotic cytokine transforming growth factor-ß (TGF-ß) has been associated with the onset and progression of the fibrosis seen in the autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc). AREA COVERED: This review explores the data supporting the notion that TGF-ß contributes to SSc fibrosis and examines why initiating clinical trials in SSc aimed at targeting integrin-mediated latent TGF-ß activation is timely. EXPERT OPINION: Targeting TGF-ß directly has not been proven to be clinically effective in this disease. Conversely, targeting matrix stiffness, which perpetuates fibrosis, may have more promise. Intriguingly, targeting integrin-mediated activation of latent TGF-ß, which bridges these concepts, may have therapeutic value.


Asunto(s)
Enfermedades Autoinmunes , Esclerodermia Sistémica , Humanos , Integrina alfaV , Factor de Crecimiento Transformador beta/uso terapéutico , Esclerodermia Sistémica/tratamiento farmacológico , Fibrosis
3.
Matrix Biol ; 128: 31-38, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38423396

RESUMEN

The largest mammalian organ, skin, consisting of a dermal connective tissue layer that underlies and supports the epidermis, acts as a protective barrier that excludes external pathogens and disseminates sensory signals emanating from the local microenvironment. Dermal connective tissue is comprised of a collagen-rich extracellular matrix (ECM) that is produced by connective tissue fibroblasts resident within the dermis. When wounded, a tissue repair program is induced whereby fibroblasts, in response to alterations in the microenvironment, produce new ECM components, resulting in the formation of a scar. Failure to terminate the normal tissue repair program causes fibrotic conditions including: hypertrophic scars, keloids, and the systemic autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc). Histological and single-cell RNA sequencing (scRNAseq) studies have revealed that fibroblasts are heterogeneous and highly plastic. Understanding how this diversity contributes to dermal homeostasis, wounding, fibrosis, and cancer may ultimately result in novel anti-fibrotic therapies and personalized medicine. This review summarizes studies supporting this concept.


Asunto(s)
Cicatriz Hipertrófica , Esclerodermia Sistémica , Animales , Epidermis/patología , Fibroblastos/patología , Fibrosis , Mamíferos , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Piel/patología
4.
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
5.
Arch Oral Biol ; 160: 105910, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38364717

RESUMEN

OBJECTIVE: To determine whether celastrol, an inhibitor of the mechanosensitive transcriptional cofactor yes-associated protein-1 (YAP1), impairs the ability of TGFß1 to stimulate fibrogenic activity in human gingival fibroblast cell line. DESIGN: Human gingival fibroblasts were pre-treated with celastrol or DMSO followed by stimulation with or without TGFß1 (4 ng/ml). We then utilized bulk RNA sequencing (RNAseq), real-time polymerase chain reaction (RT-PCR), Western blot, immunofluorescence, cell proliferation assays to determine if celastrol impaired TGFß1-induced responses in a human gingival fibroblast cell line. RESULTS: Celastrol impaired the ability of TGFß1 to induce expression of the profibrotic marker and mediator CCN2. Bulk RNAseq analysis of gingival fibroblasts treated with TGFß1, in the presence or absence of celastrol, revealed that celastrol impaired the ability of TGFß1 to induce mRNA expression of genes within extracellular matrix, wound healing, focal adhesion and cytokine/Wnt signaling clusters. RT-PCR analysis of extracted RNAs confirmed that celastrol antagonized the ability of TGFß1 to induce expression of genes anticipated to contribute to fibrotic responses. Celastrol also reduced gingival fibroblast proliferation, and YAP1 nuclear localization in response to TGFß1. CONCLUSION: YAP1 inhibitors such as celastrol could be used to impair pro-fibrotic responses to TGFß1 in human gingival fibroblasts.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Triterpenos Pentacíclicos , Factor de Crecimiento Transformador beta , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Proteínas Señalizadoras YAP , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Transcripción/metabolismo , Fibroblastos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Cultivadas
6.
Nat Rev Rheumatol ; 19(11): 713-723, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37789119

RESUMEN

Fibrosis is the excessive deposition of a stable extracellular matrix (ECM); fibrotic tissue is composed principally of highly crosslinked type I collagen and highly contractile myofibroblasts. Systemic sclerosis (SSc) is a multisystem autoimmune connective tissue disease characterized by skin and organ fibrosis. The fibrotic process has been recognized in SSc for >40 years, but drugs with demonstrable efficacy against SSc fibrosis in ameliorating the lung involvement have only recently been identified. Unfortunately, these treatments are ineffective at improving the skin score in patients with SSc. Previous clinical trials in SSc have largely focused on the cross-purposing of anti-inflammatory drugs and the use of immunosuppressive drugs from the transplantation field, which address inflammatory and/or autoimmune processes. Limited examination has taken place of specific anti-fibrotic agents developed through their ability to directly target the ECM in SSc by, for example, alleviating the persistent matrix stiffness and mechanotransduction that might be required for both the initiation and maintenance of fibrosis, including in SSc. However, because of the importance of the ECM in the SSc phenotype, attempts have now been made to identify drugs that specifically target the ECM, including some drugs that are currently under consideration for the treatment of cancer.


Asunto(s)
Enfermedades Autoinmunes , Esclerodermia Sistémica , Humanos , Mecanotransducción Celular , Esclerodermia Sistémica/genética , Fibrosis , Matriz Extracelular , Enfermedades Autoinmunes/patología , Piel/patología , Fibroblastos
7.
Cells ; 12(16)2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37626831

RESUMEN

Adult human gingival fibroblasts (HGFs), the most abundant cells in the oral cavity, are essential for maintaining oral homeostasis. Compared with other tissues, adult oral mucosal wounds heal regeneratively, without scarring. Relative to fibroblasts from other locations, HGFs are relatively refractory to myofibroblast differentiation, immunomodulatory, highly regenerative, readily obtained via minimally invasive procedures, easily and rapidly expanded in vitro, and highly responsive to growth factors and cytokines. Consequently, HGFs might be a superior, yet perhaps underappreciated, source of adult mesenchymal progenitor cells to use in tissue engineering and regeneration applications, including the treatment of fibrotic auto-immune connective tissue diseases such as scleroderma. Herein, we highlight in vitro and translational studies that have investigated the regenerative and differentiation potential of HGFs, with the objective of outlining current limitations and inspiring future research that could facilitate translating the regenerative potential of HGFs into the clinic.


Asunto(s)
Encía , Medicina Regenerativa , Adulto , Humanos , Fibroblastos , Boca , Mucosa Bucal
9.
Ann Rheum Dis ; 82(9): 1191-1204, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37328193

RESUMEN

OBJECTIVES: Systemic sclerosis (SSc) is characterised by extensive tissue fibrosis maintained by mechanotranductive/proadhesive signalling. Drugs targeting this pathway are therefore of likely therapeutic benefit. The mechanosensitive transcriptional co-activator, yes activated protein-1 (YAP1), is activated in SSc fibroblasts. The terpenoid celastrol is a YAP1 inhibitor; however, if celastrol can alleviate SSc fibrosis is unknown. Moreover, the cell niches required for skin fibrosis are unknown. METHODS: Human dermal fibroblasts from healthy individuals and patients with diffuse cutaneous SSc were treated with or without transforming growth factor ß1 (TGFß1), with or without celastrol. Mice were subjected to the bleomycin-induced model of skin SSc, in the presence or absence of celastrol. Fibrosis was assessed using RNA Sequencing, real-time PCR, spatial transcriptomic analyses, Western blot, ELISA and histological analyses. RESULTS: In dermal fibroblasts, celastrol impaired the ability of TGFß1 to induce an SSc-like pattern of gene expression, including that of cellular communication network factor 2, collagen I and TGFß1. Celastrol alleviated the persistent fibrotic phenotype of dermal fibroblasts cultured from lesions of SSc patients. In the bleomycin-induced model of skin SSc, increased expression of genes associated with reticular fibroblast and hippo/YAP clusters was observed; conversely, celastrol inhibited these bleomycin-induced changes and blocked nuclear localisation of YAP. CONCLUSIONS: Our data clarify niches within the skin activated in fibrosis and suggest that compounds, such as celastrol, that antagonise the YAP pathway may be potential treatments for SSc skin fibrosis.


Asunto(s)
Esclerodermia Sistémica , Enfermedades de la Piel , Humanos , Animales , Ratones , Tripterygium , Esclerodermia Sistémica/patología , Fibrosis , Enfermedades de la Piel/patología , Piel/patología , Bleomicina/farmacología , Fibroblastos/metabolismo , Factores de Transcripción/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad
10.
Clin Epigenetics ; 15(1): 96, 2023 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-37270501

RESUMEN

BACKGROUND: Abnormal DNA methylation is thought to contribute to the onset and progression of systemic sclerosis. Currently, the most comprehensive assay for profiling DNA methylation is whole-genome bisulfite sequencing (WGBS), but its precision depends on read depth and it may be subject to sequencing errors. SOMNiBUS, a method for regional analysis, attempts to overcome some of these limitations. Using SOMNiBUS, we re-analyzed WGBS data previously analyzed using bumphunter, an approach that initially fits single CpG associations, to contrast DNA methylation estimates by both methods. METHODS: Purified CD4+ T lymphocytes of 9 SSc and 4 control females were sequenced using WGBS. We separated the resulting sequencing data into regions with dense CpG data, and differentially methylated regions (DMRs) were inferred with the SOMNiBUS region-level test, adjusted for age. Pathway enrichment analysis was performed with ingenuity pathway analysis (IPA). We compared the results obtained by SOMNiBUS and bumphunter. RESULTS: Of 8268 CpG regions of ≥ 60 CpGs eligible for analysis with SOMNiBUS, we identified 131 DMRs and 125 differentially methylated genes (DMGs; p-values less than Bonferroni-corrected threshold of 6.05-06 controlling family-wise error rate at 0.05; 1.6% of the regions). In comparison, bumphunter identified 821,929 CpG regions, 599 DMRs (of which none had ≥ 60 CpGs) and 340 DMGs (q-value of 0.05; 0.04% of all regions). The top ranked gene identified by SOMNiBUS was FLT4, a lymphangiogenic orchestrator, and the top ranked gene on chromosome X was CHST7, known to catalyze the sulfation of glycosaminoglycans in the extracellular matrix. The top networks identified by IPA included connective tissue disorders. CONCLUSIONS: SOMNiBUS is a complementary method of analyzing WGBS data that enhances biological insights into SSc and provides novel avenues of investigation into its pathogenesis.


Asunto(s)
Metilación de ADN , Esclerodermia Sistémica , Femenino , Humanos , Islas de CpG , Secuenciación Completa del Genoma/métodos , Esclerodermia Sistémica/genética
11.
Matrix Biol ; 119: 125-140, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37080324

RESUMEN

Previously, impaired responses to immunotherapy in cancer had been attributed mainly to inherent tumor characteristics (tumor cell intrinsic factors) such as low immunogenicity, (low) mutational burden, weak host immune system, etc. However, mapping the responses of immunotherapeutic regimes in clinical trials for different types of cancer has pointed towards an obvious commonality - that tumors with a rich fibrotic stroma respond poorly or not at all. This has prompted a harder look on tumor cell extrinsic factors such as the surrounding tumor microenvironment (TME), and specifically, the fibrotic stroma as a potential enabler of immunotherapy failure. Indeed, the role of cancer-associated fibrosis in impeding efficacy of immunotherapy is now well-established. In fact, recent studies reveal a complex interconnection between fibrosis and treatment efficacy. Accordingly, in this review we provide a general overview of what a tumor associated fibrotic reaction is and how it interacts with the members of immune system that are frequently seen to be modulated in a failed immunotherapeutic regime.


Asunto(s)
Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Inmunoterapia , Fibrosis , Microambiente Tumoral
12.
JCI Insight ; 8(1)2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36625347

RESUMEN

Vascular smooth muscle cell (SMC) phenotypic switching is widely recognized as a key mechanism responsible for the pathogenesis of several aortic diseases, such as aortic aneurysm. Cellular communication network factor 2 (CCN2), often upregulated in human pathologies and animal disease models, exerts myriad context-dependent biological functions. However, current understanding of the role of SMC-CCN2 in SMC phenotypic switching and its function in the pathology of abdominal aortic aneurysm (AAA) is lacking. Here, we show that SMC-restricted CCN2 deficiency causes AAA in the infrarenal aorta of angiotensin II-infused (Ang II-infused) hypercholesterolemic mice at a similar anatomic location to human AAA. Notably, the resistance of naive C57BL/6 WT mice to Ang II-induced AAA formation is lost upon silencing of CCN2 in SMC. Furthermore, the pro-AAA phenotype of SMC-CCN2-KO mice is recapitulated in a different model that involves the application of elastase-ß-aminopropionitrile. Mechanistically, our findings reveal that CCN2 intersects with TGF-ß signaling and regulates SMC marker expression. Deficiency of CCN2 triggers SMC reprograming associated with alterations in Krüppel-like factor 4 and contractile marker expression, and this reprograming likely contributes to the development of AAA in mice. These results identify SMC-CCN2 as potentially a novel regulator of SMC phenotypic switching and AA biology.


Asunto(s)
Aneurisma de la Aorta Abdominal , Músculo Liso Vascular , Humanos , Ratones , Animales , Músculo Liso Vascular/patología , Reprogramación Celular , Ratones Endogámicos C57BL , Aneurisma de la Aorta Abdominal/metabolismo , Miocitos del Músculo Liso/metabolismo
13.
Methods Mol Biol ; 2582: 309-321, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36370359

RESUMEN

Approximately 45% of the deaths in the developed world result from conditions with a fibrotic component. Although no specific, focused anti-fibrotic therapies have been approved for clinical use, a long-standing concept is that targeting CCN proteins may be useful to treat fibrosis. Herein, we summarize current data supporting the concept that targeting CCN2 may be a viable anti-fibrotic approach to treat scleroderma. Testing this hypothesis has been made possible by using a mouse model of inflammation-driven skin and lung fibrosis.


Asunto(s)
Fibrosis Pulmonar , Esclerodermia Sistémica , Animales , Bleomicina/efectos adversos , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibrosis , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Piel/metabolismo , Modelos Animales de Enfermedad , Esclerodermia Sistémica/inducido químicamente , Esclerodermia Sistémica/metabolismo
14.
J Cell Commun Signal ; 16(4): 631-632, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35352286
15.
Matrix Biol ; 109: 49-69, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35346795

RESUMEN

The cardiac extracellular matrix plays essential roles in homeostasis and injury responses. Although the role of fibrillar collagens have been thoroughly documented, the functions of non-fibrillar collagen members remain underexplored. These include a distinct group of non-fibrillar collagens, termed, fibril-associated collagens with interrupted triple helices (FACITs). Recent reports of collagen type XIX (encoded by Col19a1) expression in adult heart and evidence of its enhanced expression in cardiac ischemia suggest important functions for this FACIT in cardiac ECM structure and function. Here, we examined the cellular source of collagen XIX in the adult murine heart and evaluated its involvement in ECM structure and ventricular function. Immunodetection of collagen XIX in fractionated cardiovascular cell lineages revealed fibroblasts and smooth muscle cells as the primary sources of collagen XIX in the heart. Based on echocardiographic and histologic analyses, Col19a1 null (Col19a1N/N) mice exhibited reduced systolic function, thinning of left ventricular walls, and increased cardiomyocyte cross-sectional areas-without gross changes in myocardial collagen content or basement membrane morphology. Col19a1N/N cardiac fibroblasts had augmented expression of several enzymes involved in the synthesis and stability of fibrillar collagens, including PLOD1 and LOX. Furthermore, second harmonic generation-imaged ECM derived from Col19a1N/N cardiac fibroblasts, and transmission electron micrographs of decellularized hearts from Col19a1N/N null animals, showed marked reductions in fibrillar collagen structural organization. Col19a1N/N mice also displayed enhanced phosphorylation of focal adhesion kinase (FAK), signifying de-repression of the FAK pathway-a critical mediator of cardiomyocyte hypertrophy. Collectively, we show that collagen XIX, which had a heretofore unknown role in the mammalian heart, participates in the regulation of cardiac structure and function-potentially through modulation of ECM fibrillar collagen structural organization. Further, these data suggest that this FACIT may modify ECM superstructure via acting at the level of the fibroblast to regulate their expression of collagen synthetic and stabilization enzymes.


Asunto(s)
Colágeno , Colágenos Asociados a Fibrillas , Animales , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Colágenos Asociados a Fibrillas/metabolismo , Colágenos Fibrilares/metabolismo , Mamíferos/metabolismo , Ratones , Función Ventricular
16.
J Cell Commun Signal ; 15(4): 473, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34292484
17.
J Cell Commun Signal ; 15(3): 465-466, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34014499

RESUMEN

Mesenchymal progenitor cells play a key role in fibrogenesis. An exciting paper was recently published showed that blister fluid from the skin patients with the autoimmune connective tissue disease scleroderma (systemic sclerosis, SSc) preferentially activated mesenchymal progenitor cells (Taki et al. in Arthritis Rheumatol 72(8):1361-1374, 2020). These data provide new and invaluable insights into the complex interactions in the connective tissue microenvironment that ultimately result in persistent, pathological fibrosis.

18.
Neoplasia ; 23(4): 375-390, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33784590

RESUMEN

The tumor microenvironment (TME) is an important mediator of breast cancer progression. Cancer-associated fibroblasts constitute a major component of the TME and may originate from tissue-associated fibroblasts or infiltrating mesenchymal stromal cells (MSCs). The mechanisms by which cancer cells activate fibroblasts and recruit MSCs to the TME are largely unknown, but likely include deposition of a pro-tumorigenic secretome. The secreted embryonic protein NODAL is clinically associated with breast cancer stage and promotes tumor growth, metastasis, and vascularization. Herein, we show that NODAL expression correlates with the presence of activated fibroblasts in human triple-negative breast cancers and that it directly induces Cancer-associated fibroblasts phenotypes. We further show that NODAL reprograms cancer cell secretomes by simultaneously altering levels of chemokines (e.g., CXCL1), cytokines (e.g., IL-6) and growth factors (e.g., PDGFRA), leading to alterations in MSC chemotaxis. We therefore demonstrate a hitherto unappreciated mechanism underlying the dynamic regulation of the TME.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína Nodal/genética , Proteína Nodal/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/fisiología , Actinas/metabolismo , Línea Celular Tumoral , Quimiocina CXCL1/metabolismo , Quimiotaxis/fisiología , Femenino , Humanos , Interleucina-6/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Neoplasias de la Mama Triple Negativas/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
J Cell Commun Signal ; 15(1): 5, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33492622
20.
J Cell Commun Signal ; 15(1): 71-80, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33398723

RESUMEN

Fibrosis is perpetuated by an autocrine, pro-adhesive signaling loop maintained by the synthetic and contractile abilities of myofibroblasts and the stiff, highly-crosslinked extracellular matrix. Transcriptional complexes that are exquisitely responsive to mechanotransduction include the co-activator YAP1, which regulates the expression of members of the CCN family of matricellular proteins such as CCN2 and CCN1. Although selective YAP1 inhibitors exist, the effect of these inhibitors on profibrotic gene expression in fibroblasts is largely unknown, and is the subject of our current study. Herein, we use genome-wide expression profiling, real-time polymerase chain reaction and Western blot analyses, cell migration and collagen gel contraction assays to assess the ability of a selective YAP inhibitor verteporfin (VP) to block fibrogenic activities in dermal fibroblasts from healthy individual human controls and those from isolated from fibrotic lesions of patients with diffuse cutaneous systemic sclerosis (dcSSc). In control fibroblasts, VP selectively reduced expression of fibrogenic genes and also blocked the ability of TGFbeta to induce actin stress fibers in dermal fibroblasts. VP also reduced the persistent profibrotic phenotype of dermal fibroblasts cultured from fibrotic lesions of patients with dcSSc. Our results are consistent with the notion that, in the future, YAP1 inhibitors may represent a novel, valuable method of treating fibrosis as seen in dcSSc.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...