Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Matrix Biol ; 133: 103-115, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39153517

ABSTRACT

Skeletal muscle fibrosis is defined as the excessive accumulation of extracellular matrix (ECM) components and is a hallmark of muscular dystrophies. Fibro-adipogenic progenitors (FAPs) are the main source of ECM, and thus have been strongly implicated in fibrogenesis. In skeletal muscle fibrotic models, including muscular dystrophies, FAPs undergo dysregulations in terms of proliferation, differentiation, and apoptosis, however few studies have explored the impact of FAPs migration. Here, we studied fibroblast and FAPs migration and identified lysophosphatidic acid (LPA), a signaling lipid central to skeletal muscle fibrogenesis, as a significant migration inductor. We identified LPA receptor 1 (LPA1) mediated signaling as crucial for this effect through a mechanism dependent on the Hippo pathway, another pathway implicated in fibrosis across diverse tissues. This cross-talk favors the activation of the Yes-associated protein 1 (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ), leading to increased expression of fibrosis-associated genes. This study reveals the role of YAP in LPA-mediated fibrotic responses as inhibition of YAP transcriptional coactivator activity hinders LPA-induced migration in fibroblasts and FAPs. Moreover, we found that FAPs derived from the mdx4cv mice, a murine model of Duchenne muscular dystrophy, display a heightened migratory phenotype due to enhanced LPA signaling compared to wild-type FAPs. Remarkably, we found that the inhibition of LPA1 or YAP transcriptional coactivator activity in mdx4cv FAPs reverts this phenotype. In summary, the identified LPA-LPA1-YAP pathway emerges as a critical driver of skeletal muscle FAPs migration and provides insights into potential novel targets to mitigate fibrosis in muscular dystrophies.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Movement , Fibroblasts , Fibrosis , Lysophospholipids , Muscle, Skeletal , Receptors, Lysophosphatidic Acid , Signal Transduction , YAP-Signaling Proteins , Lysophospholipids/metabolism , Animals , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Mice , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Hippo Signaling Pathway , Mice, Inbred mdx , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Adipogenesis/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology
2.
Matrix Biol ; 130: 36-46, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723870

ABSTRACT

Cellular Communication Network Factor 2, CCN2, is a profibrotic cytokine implicated in physiological and pathological processes in mammals. The expression of CCN2 is markedly increased in dystrophic muscles. Interestingly, diminishing CCN2 genetically or inhibiting its function improves the phenotypes of chronic muscular fibrosis in rodent models. Elucidating the cell-specific mechanisms behind the induction of CCN2 is a fundamental step in understanding its relevance in muscular dystrophies. Here, we show that the small lipids LPA and 2S-OMPT induce CCN2 expression in fibro/adipogenic progenitors (FAPs) through the activation of the LPA1 receptor and, to a lower extent, by also the LPA6 receptor. These cells show a stronger induction than myoblasts or myotubes. We show that the LPA/LPARs axis requires ROCK kinase activity and organized actin cytoskeleton upstream of YAP/TAZ signaling effectors to upregulate CCN2 levels, suggesting that mechanical signals are part of the mechanism behind this process. In conclusion, we explored the role of the LPA/LPAR axis on CCN2 expression, showing a strong cytoskeletal-dependent response in muscular FAPs.


Subject(s)
Adipogenesis , Connective Tissue Growth Factor , Lysophospholipids , Animals , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Mice , Lysophospholipids/metabolism , Cell Communication , Signal Transduction , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Stem Cells/metabolism , Stem Cells/cytology , Gene Expression Regulation , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Cell Differentiation , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Humans , Actin Cytoskeleton/metabolism
3.
Matrix Biol ; 119: 57-81, 2023 05.
Article in English | MEDLINE | ID: mdl-37137584

ABSTRACT

Lysophosphatidic acid (LPA) is a lysophospholipid that signals through six G-protein coupled receptors (LPARs), LPA1 to LPA6. LPA has been described as a potent modulator of fibrosis in different pathologies. In skeletal muscle, LPA increases fibrosis-related proteins and the number of fibro/adipogenic progenitors (FAPs). FAPs are the primary source of ECM-secreting myofibroblasts in acute and chronic damage. However, the effect of LPA on FAPs activation in vitro has not been explored. This study aimed to investigate FAPs' response to LPA and the downstream signaling mediators involved. Here, we demonstrated that LPA mediates FAPs activation by increasing their proliferation, expression of myofibroblasts markers, and upregulation of fibrosis-related proteins. Pretreatment with the LPA1/LPA3 antagonist Ki16425 or genetic deletion of LPA1 attenuated the LPA-induced FAPs activation, resulting in decreased expression of cyclin e1, α-SMA, and fibronectin. We also evaluated the activation of the focal adhesion kinase (FAK) in response to LPA. Our results showed that LPA induces FAK phosphorylation in FAPs. Treatment with the P-FAK inhibitor PF-228 partially prevented the induction of cell responses involved in FAPs activation, suggesting that this pathway mediates LPA signaling. FAK activation controls downstream cell signaling within the cytoplasm, such as the Hippo pathway. LPA induced the dephosphorylation of the transcriptional coactivator YAP (Yes-associated protein) and promoted direct expression of target pathway genes such as Ctgf/Ccn2 and Ccn1. The blockage of YAP transcriptional activity with Super-TDU further confirmed the role of YAP in LPA-induced FAPs activation. Finally, we demonstrated that FAK is required for LPA-dependent YAP dephosphorylation and the induction of Hippo pathway target genes. In conclusion, LPA signals through LPA1 to regulate FAPs activation by activating FAK to control the Hippo pathway.


Subject(s)
Hippo Signaling Pathway , Lysophospholipids , Humans , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Lysophospholipids/pharmacology , Lysophospholipids/metabolism , Muscle, Skeletal/metabolism , Fibrosis
4.
Matrix Biol ; 82: 20-37, 2019 09.
Article in English | MEDLINE | ID: mdl-30716392

ABSTRACT

Muscular fibrosis is caused by excessive accumulation of extracellular matrix (ECM) that replaces functional tissue, and it is a feature of several myopathies and neuropathies. Knowledge of the biology and regulation of pro-fibrotic factors is critical for the development of new therapeutic strategies. Upon unilateral sciatic nerve transection, we observed accumulation of ECM proteins such as collagen and fibronectin in the denervated hindlimb, together with increased levels of the profibrotic factors transforming growth factor type ß (TGF-ß) and connective tissue growth factor (CTGF/CCN2). In mice hemizygous for CTGF/CCN2 or in mice treated with a blocking antibody against CTGF/CCN2, we observed reduced accumulation of ECM proteins after denervation as compared to control mice, with no changes in fibro/adipogenic progenitors (FAPs), suggesting a direct role of CTGF/CCN2 on denervation-induced fibrosis. During time course experiments, we observed that ECM proteins and CTGF/CCN2 levels are increased early after denervation (2-4 days), while TGF-ß signaling shows a delayed kinetics of appearance (1-2 weeks). Furthermore, blockade of TGF-ß signaling does not decrease fibronectin or CTGF levels after 4 days of denervation. These results suggest that in our model CTGF/CCN2 is not up-regulated by canonical TGF-ß signaling early after denervation and that other factors are likely involved in the early fibrotic response following skeletal muscle denervation.


Subject(s)
Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Benzamides/pharmacology , Connective Tissue Growth Factor/antagonists & inhibitors , Dioxoles/pharmacology , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Regulation , Imidazoles/pharmacology , Male , Mice , Models, Animal , Muscle Denervation , Muscle, Skeletal/metabolism , Quinoxalines/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL