Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.448
Filtrer
1.
F S Sci ; 5(1): 16-23, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-39382269

RÉSUMÉ

OBJECTIVE: To investigate the direct effect of growth differentiation factor 9 (GDF9) on androgen production in human theca cells. DESIGN: Experimental study. SETTING: Tertiary hospital-based research laboratory. PATIENT(S): Women who underwent in vitro fertilization and intracytoplasmic sperm injections at our clinic were included in this study. INTERVENTION(S): Primary cultured human theca cells from women undergoing in vitro fertilization and intracytoplasmic sperm injection treatment were treated with GDF9, an activin receptor-like kinase 5 (ALK5) inhibitor, and a SMAD4 agonist. MAIN OUTCOME MEASURE(S): The expression of androgen synthesis-related genes StAR, CYP17A1, and LHCGR, levels of androstenedione and testosterone, phosphorylation of SMAD2/3, and the interaction between bone morphogenic protein-activated type II receptor and ALK5 were evaluated using reverse transcription-quantitative polymerase chain reaction, Western blot, enzyme-linked immunosorbent assays, and coimmunoprecipitation assays, respectively. RESULT(S): Growth differentiation factor 9 decreased StAR, CYP17A1, and LHCGR expression levels in human theca cells, which was prevented by treatment with the ALK5 inhibitor, and suppressed production of androgen in human theca cells. Growth differentiation factor 9 increased SMAD2/3 phosphorylation, and the ALK5 inhibitor also suppressed this effect. Bone morphogenic protein-activated type II receptor and ALK5 bound to each other after GDF9 stimulation. The SMAD4 agonist kartogenin also decreased messenger RNA levels of StAR and CYP17A1 and protein levels of StAR in human theca cells. CONCLUSION(S): Growth differentiation factor 9 can activate the bone morphogenic protein-activated type II receptor-ALK5-SMAD2/3 signaling pathway, suppress CYP17A1 expression, and decrease androgen production in human theca cells.


Sujet(s)
Facteur-9 de croissance et de différenciation , Récepteur de type I du facteur de croissance transformant bêta , Récepteurs TGF-bêta , Protéine Smad2 , Protéine Smad-3 , Steroid 17-alpha-hydroxylase , Cellules thécales , Humains , Steroid 17-alpha-hydroxylase/génétique , Steroid 17-alpha-hydroxylase/métabolisme , Cellules thécales/métabolisme , Cellules thécales/effets des médicaments et des substances chimiques , Femelle , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/génétique , Facteur-9 de croissance et de différenciation/métabolisme , Facteur-9 de croissance et de différenciation/génétique , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Récepteur de type I du facteur de croissance transformant bêta/génétique , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Androgènes/métabolisme , Récepteur LH/génétique , Récepteur LH/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Phosphoprotéines/métabolisme , Phosphoprotéines/génétique , Protéine Smad-4/métabolisme , Protéine Smad-4/génétique , Phosphorylation/effets des médicaments et des substances chimiques , Cellules cultivées , Ovocytes/métabolisme , Ovocytes/effets des médicaments et des substances chimiques , Androstènedione/métabolisme , Testostérone/métabolisme , Récepteur de type II du facteur de croissance transformant bêta/génétique , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
2.
Front Endocrinol (Lausanne) ; 15: 1468364, 2024.
Article de Anglais | MEDLINE | ID: mdl-39351532

RÉSUMÉ

Anti-Müllerian hormone (AMH) is a peptide belonging to the transforming growth factor beta superfamily and acts exclusively through its receptor type 2 (AMHR2). From the 8th week of pregnancy, AMH is produced by Sertoli cells, and from the 23rd week of gestation, it is produced by granulosa cells of the ovary. AMH plays a critical role in regulating gonadotropin secretion, ovarian tissue responsiveness to pituitary hormones, and the pathogenesis of polycystic ovarian syndrome. It inhibits the transition from primordial to primary follicles and is considered the best marker of ovarian reserve. Therefore, measuring AMH concentration of the hormone is valuable in managing assisted reproductive technologies. AMH was initially discovered through its role in the degeneration of Müllerian ducts in male fetuses. However, due to its ability to inhibit the cell cycle and induce apoptosis, it has also garnered interest in oncology. For example, antibodies targeting AMHR2 are being investigated for their potential in diagnosing and treating various cancers. Additionally, AMH is present in motor neurons and functions as a protective and growth factor. Consequently, it is involved in learning and memory processes and may support the treatment of Alzheimer's disease. This review aims to provide a comprehensive overview of the biology of AMH and its role in both endocrinology and oncology.


Sujet(s)
Hormone antimullérienne , Tumeurs , Hormone antimullérienne/métabolisme , Humains , Tumeurs/métabolisme , Femelle , Animaux , Récepteurs TGF-bêta/métabolisme , Récepteurs peptidiques/métabolisme , Mâle , Endocrinologie/tendances , Endocrinologie/méthodes
3.
Biomolecules ; 14(9)2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39334892

RÉSUMÉ

Background: Esophageal organoids from a variety of pathologies including cancer are grown in Advanced Dulbecco's Modified Eagle Medium-Nutrient Mixture F12 (hereafter ADF). However, the currently available ADF-based formulations are suboptimal for normal human esophageal organoids, limiting the ability to compare normal esophageal organoids with those representing a given disease state. Methods: We have utilized immortalized normal human esophageal epithelial cell (keratinocyte) lines EPC1 and EPC2 and endoscopic normal esophageal biopsies to generate three-dimensional (3D) organoids. To optimize the ADF-based medium, we evaluated the requirement of exogenous epidermal growth factor (EGF) and inhibition of transforming growth factor-(TGF)-ß receptor-mediated signaling, both key regulators of the proliferation of human esophageal keratinocytes. We have modeled human esophageal epithelial pathology by stimulating esophageal 3D organoids with interleukin (IL)-13, an inflammatory cytokine, or UAB30, a novel pharmacological activator of retinoic acid signaling. Results: The formation of normal human esophageal 3D organoids was limited by excessive EGF and intrinsic TGFß-receptor-mediated signaling. Optimized HOME0 improved normal human esophageal organoid formation. In the HOME0-grown organoids, IL-13 and UAB30 induced epithelial changes reminiscent of basal cell hyperplasia, a common histopathologic feature in broad esophageal disease conditions including eosinophilic esophagitis. Conclusions: HOME0 allows modeling of the homeostatic differentiation gradient and perturbation of the human esophageal epithelium while permitting a comparison of organoids from mice and other organs grown in ADF-based media.


Sujet(s)
Oesophage , Homéostasie , Organoïdes , Humains , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/métabolisme , Oesophage/métabolisme , Oesophage/anatomopathologie , Oesophage/effets des médicaments et des substances chimiques , Facteur de croissance épidermique/pharmacologie , Facteur de croissance épidermique/métabolisme , Kératinocytes/métabolisme , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/cytologie , Transduction du signal/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Modèles biologiques , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteurs TGF-bêta/métabolisme
4.
Cell Death Dis ; 15(9): 654, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39231945

RÉSUMÉ

Transforming growth factor (TGF)-ß signaling is critical for epithelial-mesenchymal transition (EMT) and colorectal cancer (CRC) metastasis. Disruption of Smad-depednent TGF-ß signaling has been shown in CRC cells. However, TGF-ß receptor remains expressed on CRC cells. Here, we investigated whether the cooperation between tumor-associated N-glycosylation and a glycan-binding protein modulated the TGF-ß-driven signaling and metastasis of CRC. We showed that galectin-8, a galactose-binding lectin, hampered TGF-ß-induced EMT by interacting with the type II TGF-ß receptor and competing with TGF-ß binding. Depletion of galectin-8 promoted the migration of CRC cells by increasing TGF-ß-receptor-mediated RAS and Src signaling, which was attenuated after recombinant galectin-8 treatment. Treatment with recombinant galectin-8 also induces JNK-dependent apoptosis in CRC cells. The anti-migratory effect of galectin-8 depended on ß4-galactosyltransferase-I (B4GALT1), an enzyme involved in N-glycan synthesis. Increased B4GALT1 expression was observed in clinical CRC samples. Depletion of B4GALT1 reduced the metastatic potential of CRC cells. Furthermore, inducible expression of galectin-8 attenuated tumor development and metastasis of CRC cells in an intra-splenic injection model. Our results thus demonstrate that galectin-8 alters non-canonical TGF-ß response in CRC cells and suppresses CRC progression.


Sujet(s)
Mouvement cellulaire , Tumeurs colorectales , Transition épithélio-mésenchymateuse , Galactosyltransferases , Galectines , Métastase tumorale , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Galectines/métabolisme , Galectines/génétique , Galactosyltransferases/métabolisme , Galactosyltransferases/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Évolution de la maladie , Lignée cellulaire tumorale , Transduction du signal , Souris , Récepteurs TGF-bêta/métabolisme , Souris nude , Liaison aux protéines , Apoptose/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/pharmacologie , Souris de lignée BALB C
5.
Stroke ; 55(9): 2340-2352, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39129597

RÉSUMÉ

BACKGROUND: TGF (transforming growth factor)-ß pathway is central to blood-brain barrier development as it regulates cross talk between pericytes and endothelial cells. Murine embryos lacking TGFß receptor Alk5 (activin receptor-like kinase 5) in brain pericytes (mutants) display endothelial cell hyperproliferation, abnormal vessel morphology, and gross germinal matrix hemorrhage-intraventricular hemorrhage (GMH-IVH), leading to perinatal lethality. Mechanisms underlying how ALK5 signaling in pericytes noncell autonomously regulates endothelial cell behavior remain elusive. METHODS: Transcriptomic analysis of human brain pericytes with ALK5 silencing identified differential gene expression. Brain vascular cells isolated from mutant embryonic mice with GMH-IVH and preterm human IVH brain samples were utilized for target validation. Finally, pharmacological and genetic inhibition was used to study the therapeutic effects on GMH-IVH pathology. RESULTS: Herein, we establish that the TGFß/ALK5 pathway robustly represses ANGPT2 (angiopoietin-2) in pericytes via epigenetic remodeling. TGFß-driven SMAD (suppressor of mothers against decapentaplegic) 3/4 associates with TGIF1 (TGFß-induced factor homeobox 1) and HDAC (histone deacetylase) 5 to form a corepressor complex at the Angpt2 promoter, resulting in promoter deacetylation and gene repression. Moreover, murine and human germinal matrix vessels display increased ANGPT2 expression during GMH-IVH. Isolation of vascular cells from murine germinal matrix identifies pericytes as a cellular source of excessive ANGPT2. In addition, mutant endothelial cells exhibit higher phosphorylated TIE2 (tyrosine protein kinase receptor). Pharmacological or genetic inhibition of ANGPT2 in mutants improves germinal matrix vessel morphology and attenuates GMH pathogenesis. Importantly, genetic ablation of Angpt2 in mutant pericytes prevents perinatal lethality, prolonging survival. CONCLUSIONS: This study demonstrates that TGFß-mediated ANGPT2 repression in pericytes is critical for maintaining blood-brain barrier integrity and identifies pericyte-derived ANGPT2 as an important pathological target for GMH-IVH.


Sujet(s)
Angiopoïétine-2 , Péricytes , Facteur de croissance transformant bêta , Péricytes/métabolisme , Péricytes/anatomopathologie , Animaux , Souris , Humains , Angiopoïétine-2/métabolisme , Angiopoïétine-2/génétique , Facteur de croissance transformant bêta/métabolisme , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Récepteur de type I du facteur de croissance transformant bêta/génétique , Hémorragie cérébrale/métabolisme , Hémorragie cérébrale/anatomopathologie , Hémorragie cérébrale/génétique , Transduction du signal/physiologie , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/génétique , Cellules endothéliales/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme
6.
Genesis ; 62(4): e23614, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39139086

RÉSUMÉ

Organisms from the five kingdoms of life use minerals to harden their tissues and make teeth, shells and skeletons, in the process of biomineralization. The sea urchin larval skeleton is an excellent system to study the biological regulation of biomineralization and its evolution. The gene regulatory network (GRN) that controls sea urchin skeletogenesis is known in great details and shows similarity to the GRN that controls vertebrates' vascularization while it is quite distinct from the GRN that drives vertebrates' bone formation. Yet, transforming growth factor beta (TGF-ß) signaling regulates both sea urchin and vertebrates' skeletogenesis. Here, we study the upstream regulation and identify transcriptional targets of TGF-ß in the Mediterranean Sea urchin species, Paracentrotus lividus. TGF-ßRII is transiently active in the skeletogenic cells downstream of vascular endothelial growth factor (VEGF) signaling, in P. lividus. Continuous perturbation of TGF-ßRII activity significantly impairs skeletal elongation and the expression of key skeletogenic genes. Perturbation of TGF-ßRII after skeletal initiation leads to a delay in skeletal elongation and minor changes in gene expression. TGF-ß targets are distinct from its transcriptional targets during vertebrates' bone formation, suggesting that the role of TGF-ß in biomineralization in these two phyla results from convergent evolution.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Larve , Paracentrotus , Animaux , Larve/croissance et développement , Larve/métabolisme , Larve/génétique , Paracentrotus/génétique , Paracentrotus/métabolisme , Paracentrotus/embryologie , Récepteur de type II du facteur de croissance transformant bêta/génétique , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Récepteurs TGF-bêta/génétique , Récepteurs TGF-bêta/métabolisme , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Ostéogenèse/génétique , Réseaux de régulation génique , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme
7.
Int J Biol Sci ; 20(9): 3557-3569, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993575

RÉSUMÉ

To investigate the cell linkage between tooth dentin and bones, we studied TGF-ß roles during postnatal dentin development using TGF-ß receptor 2 (Tgfßr2) cKO models and cell lineage tracing approaches. Micro-CT showed that the early Tgfßr2 cKO exhibit short roots and thin root dentin (n = 4; p<0.01), a switch from multilayer pre-odontoblasts/odontoblasts to a single-layer of bone-like cells with a significant loss of ~85% of dentinal tubules (n = 4; p<0.01), and a matrix shift from dentin to bone. Mechanistic studies revealed a statistically significant decrease in odontogenic markers, and a sharp increase in bone markers. The late Tgfßr2 cKO teeth displayed losses of odontoblast polarity, a significant reduction in crown dentin volume, and the onset of massive bone-like structures in the crown pulp with high expression levels of bone markers and low levels of dentin markers. We thus concluded that bones and tooth dentin are in the same evolutionary linkage in which TGF-ß signaling defines the odontogenic fate of dental mesenchymal cells and odontoblasts. This finding also raises the possibility of switching the pulp odontogenic to the osteogenic feature of pulp cells via a local manipulation of gene programs in future treatment of tooth fractures.


Sujet(s)
Dentine , Odontoblastes , Récepteurs TGF-bêta , Transduction du signal , Facteur de croissance transformant bêta , Dentine/métabolisme , Facteur de croissance transformant bêta/métabolisme , Animaux , Odontoblastes/métabolisme , Récepteurs TGF-bêta/métabolisme , Souris , Dent/métabolisme , Os et tissu osseux/métabolisme , Microtomographie aux rayons X , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Récepteur de type II du facteur de croissance transformant bêta/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Souris knockout
8.
Genes (Basel) ; 15(7)2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39062724

RÉSUMÉ

TGF-ß1/Smads is a classic signaling pathway, which plays important roles in the development process of organisms. Black porgy Acanthopagrus schlegelii and red porgy Pagrus major are valuable economic fishes, and their hybrid offspring show excellent heterosis traits. Yet the molecular regulation mechanism of the heterosis traits is less clear. Here, we explored the TGF-ß1/Smads pathway's molecular genetic information for heterosis in A. schlegelii ♂ × P. major ♀ (AP) and A. schlegelii ♀ × P. major ♂ (PA) in terms of growth and development. The mRNA expression levels of TGF-ß1, TßR-I, TßR-II, and Smad2 genes in different developmental stages of A. schlegelii were detected. Furthermore, the expression levels of TGF-ß1, TßR-I, TßR-II, and Smad2 genes in different tissues of adult (mRNA level) and larva (mRNA and protein level) of A. schlegelii, P. major, and their hybrids were determined by both real-time quantitative PCR and Western blot techniques. The results indicated the ubiquitous expression of these genes in all developmental stages of A. schlegelii and in all tested tissues of A. schlegelii, P. major, and its hybrids. Among them, the mRNA of TGF-ß1, TßR-I, and TßR-II genes is highly expressed in the liver, gill, kidney, and muscle of black porgy, red porgy, and their hybrid offspring. There are significant changes in gene and protein expression levels in hybrid offspring, which indirectly reflect hybrid advantage. In addition, there was no correlation between protein and mRNA expression levels of Smad2 protein. The results provide novel data for the differential expression of growth and development genes between the reciprocal hybridization generation of black porgy and red porgy and its parents, which is conducive to further explaining the molecular regulation mechanism of heterosis in the growth and development of hybrid porgy.


Sujet(s)
Vigueur hybride , Protéine Smad2 , Facteur de croissance transformant bêta-1 , Animaux , Protéine Smad2/génétique , Protéine Smad2/métabolisme , Vigueur hybride/génétique , Facteur de croissance transformant bêta-1/génétique , Facteur de croissance transformant bêta-1/métabolisme , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Hybridation génétique , Récepteur de type I du facteur de croissance transformant bêta/génétique , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Perciformes/génétique , Perciformes/croissance et développement , Perciformes/métabolisme , Récepteur de type II du facteur de croissance transformant bêta/génétique , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Récepteurs TGF-bêta/génétique , Récepteurs TGF-bêta/métabolisme , Femelle , Mâle , Régulation de l'expression des gènes au cours du développement
9.
Front Immunol ; 15: 1386260, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975349

RÉSUMÉ

Introduction: Lrba is a cytoplasmic protein involved in vesicular trafficking. Lrba-deficient (Lrba-/-) mice exhibit substantially higher levels of IgA in both serum and feces than wild-type (WT) mice. Transforming growth factor ß1 (TGFß1) and its receptors (TGFßR I and II) is essential for differentiating IgA+ B cells. Furthermore, increased IgA production suggests a potential connection between Lrba and the TGFßR signaling pathway in IgA production. However, the specific function of Lrba in B cell biology remains unknown. Aim: Given the increased IgA levels in Lrba-/- mice, the goal in this work was to explore the lymph organs where the switch to IgA occurs, and if TGFßR function is affected. Methods: Non-immunized Lrba-/- mice were compared with Lrba+/+ mice. IgA levels in the serum and feces, as well as during peripheral B cell development, were determined. IgA+ B cells and plasma cells were assessed in the small intestine and secondary lymphoid organs, such as the spleen, mesenteric lymph nodes, and Peyer's patches. The TGFßR signaling pathway was evaluated by determining the expression of TGFßR on B cells. Additionally, SMAD2 phosphorylation was measured under basal conditions and in response to recombinant TGFß. Finally, confocal microscopy was performed to investigate a possible interaction between Lrba and TGFßR in B cells. Results: Lrba-/- mice exhibited significantly higher levels of circulating IgA, IgA+ B, and plasma cells than in peripheral lymphoid organs those in WT mice. TGFßR expression on the membrane of B cells was similar in both Lrba-/- and Lrba+/+ mice. However, intracellular TGFßR expression was reduced in Lrba-/- mice. SMAD2 phosphorylation showed increased levels under basal conditions; stimulation with recombinant TGFß elicited a poorer response than in that in Lrba+/+ B cells. Finally, we found that Lrba colocalizes with TGFßR in B cells. Conclusion: Lrba is essential in controlling TGFßR signaling, subsequently regulating SMAD2 phosphorylation on B cells. This mechanism may explain the increased differentiation of IgA+ B cells and production of IgA-producing plasma cells.


Sujet(s)
Lymphocytes B , Différenciation cellulaire , Immunoglobuline A , Transduction du signal , Animaux , Souris , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Différenciation cellulaire/immunologie , Immunoglobuline A/immunologie , Souris de lignée C57BL , Souris knockout , Plaques de Peyer/immunologie , Plaques de Peyer/métabolisme , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/génétique , Protéine Smad2/métabolisme , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/métabolisme
10.
F1000Res ; 13: 120, 2024.
Article de Anglais | MEDLINE | ID: mdl-38988879

RÉSUMÉ

Fibroblasts are cells of mesenchymal origin that are found throughout the body. While these cells have several functions, their integral roles include maintaining tissue architecture through the production of key extracellular matrix components, and participation in wound healing after injury. Fibroblasts are also key mediators in disease progression during fibrosis, cancer, and other inflammatory diseases. Under these perturbed states, fibroblasts can activate into inflammatory fibroblasts or contractile myofibroblasts. Fibroblasts require various growth factors and mitogenic molecules for survival, proliferation, and differentiation. While the activity of mitogenic growth factors on fibroblasts in vitro was characterized as early as the 1970s, the proliferation and differentiation effects of growth factors on these cells in vivo are unclear. Recent work exploring the heterogeneity of fibroblasts raises questions as to whether all fibroblast cell states exhibit the same growth factor requirements. Here, we will examine and review existing studies on the influence of fibroblast growth factor receptors (FGFRs), platelet-derived growth factor receptors (PDGFRs), and transforming growth factor ß receptor (TGFßR) on fibroblast cell states.


Sujet(s)
Fibroblastes , Homéostasie , Récepteur facteur croissance fibroblaste , Récepteurs aux facteurs de croissance dérivés des plaquettes , Humains , Fibroblastes/métabolisme , Récepteurs aux facteurs de croissance dérivés des plaquettes/métabolisme , Animaux , Récepteur facteur croissance fibroblaste/métabolisme , Récepteurs TGF-bêta/métabolisme
11.
Acta Biochim Pol ; 71: 12993, 2024.
Article de Anglais | MEDLINE | ID: mdl-38983797

RÉSUMÉ

Endogenous electric fields (EFs) serve as a crucial signal to guide cell movement in processes such as wound healing, embryonic development, and cancer metastasis. However, the mechanism underlying cell electrotaxis remains poorly understood. A plausible hypothesis suggests that electrophoretic or electroosmotic forces may rearrange charged components of the cell membrane, including receptors for chemoattractants which induce asymmetric signaling and directional motility. This study aimed to explore the role of Transforming Growth Factor Beta (TGFß) signaling in the electrotactic reaction of 3T3 fibroblasts. Our findings indicate that inhibiting canonical and several non-canonical signaling pathways originating from the activated TGF-ß receptor does not hinder the directed migration of 3T3 cells to the cathode. Furthermore, suppression of TGF-ß receptor expression does not eliminate the directional migration effect of 3T3 cells in the electric field. Additionally, there is no observed redistribution of the TGF-ß receptor in the electric field. However, our studies affirm the significant involvement of Phosphoinositide 3-Kinase (PI3K) in electrotaxis, suggesting that in our model, its activation is likely associated with factors independent of TGFß action.


Sujet(s)
Mouvement cellulaire , Fibroblastes , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Souris , Facteur de croissance transformant bêta/métabolisme , Fibroblastes/métabolisme , Récepteurs TGF-bêta/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Cellules 3T3
12.
Mol Nutr Food Res ; 68(14): e2300777, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38880772

RÉSUMÉ

Organisms maintain their cellular homeostatic balance by interacting with their environment through the use of their cell surface receptors. Membrane based receptors such as the transforming growth factor ß receptor (TGFR), the prolactin receptor (PRLR), and hepatocyte growth factor receptor (HGFR), along with their associated signaling cascade, play significant roles in retaining cellular homeostasis. While these receptors and related signaling pathways are essential for health of cell and organism, their dysregulation can lead to imbalance in cell function with severe pathological conditions such as cell death or cancer. Ochratoxin A (OTA) can disrupt cellular homeostasis by altering expression levels of these receptors and/or receptor-associated intracellular downstream signaling modulators and/or pattern and levels of their phosphorylation/dephosphorylation. Recent studies have shown that the activity of the TGFR, the PRLR, and HGFR and their associated signaling cascades change upon OTA exposure. A critical evaluation of these findings suggests that while increased activity of the HGFR and TGFR signaling pathways leads to an increase in cell survival and fibrosis, decreased activity of the PRLR signaling pathway leads to tissue damage. This review explores the roles of these receptors in OTA-related pathologies and effects on cellular homeostasis.


Sujet(s)
Homéostasie , Ochratoxines , Récepteur prolactine , Transduction du signal , Ochratoxines/toxicité , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Animaux , Récepteur prolactine/métabolisme , Protéines proto-oncogènes c-met/métabolisme , Récepteurs TGF-bêta/métabolisme
13.
Cell Biochem Biophys ; 82(2): 1109-1120, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38834831

RÉSUMÉ

BACKGROUND: Rho-kinase (ROCK) regulates actomyosin contraction, coronary vasospasm, and cytoskeleton dynamics. ROCK and of NADPH oxidase (NOX) play an essential role in cardiovascular disease and proteoglycan synthesis, which promotes atherosclerosis by trapping low density lipoprotein. ROCK is activated by endothelin-1 (ET1) and transactivates the transforming growth factor beta receptor (TGFßR1), intensifying Smad signaling and proteoglycan production. This study aimed to identify the role of myosin light chain phosphatase (MLCP) as a downstream target of ROCK in TßR1 transactivation. METHODS: Vascular smooth muscle cells were treated with ET1 and inhibitors of ROCK and MLCP were added. The phosphorylation levels of Smad2C, myosin light chain (MLC), and MLCP were monitored by western blot, and the mRNA expression of chondroitin 4-O-sulfotransferase 1 (C4ST1) was assessed by quantitative real-time PCR. RESULTS: We examined ROCK's role in ET1-induced TGFßR1 activation. ROCK phosphorylated MLCP at the MYPT1 T853 residue, blocked by the ROCK inhibitor Y27632. ROCK also increased MLC phosphorylation and actomyosin contraction in response to ET1, enhanced by the phosphatase inhibitor Calyculin A. Calyculin A also increased C4ST1 expression, GAG-chain synthesizing enzymes. CONCLUSIONS: This work suggests that ROCK is involved in ET1-mediated TßR1 activation through increased MLCP phosphorylation, which leads to Smad2C phosphorylation and stimulates C4ST1 expression.


Sujet(s)
Endothéline-1 , Myosin-light-chain phosphatase , Activation de la transcription , rho-Associated Kinases , Animaux , Humains , Amides/pharmacologie , Endothéline-1/métabolisme , Toxines de la flore et de la faune marines , Muscles lisses vasculaires/cytologie , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myosin-light-chain phosphatase/métabolisme , Myosin-light-chain phosphatase/génétique , Oxazoles , Phosphorylation/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Récepteurs TGF-bêta/métabolisme , rho-Associated Kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéine Smad2/métabolisme , Activation de la transcription/effets des médicaments et des substances chimiques
14.
Reprod Biomed Online ; 49(2): 103981, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38870625

RÉSUMÉ

RESEARCH QUESTION: What is the involvement of pigment epithelium-derived factor (PEDF), expressed in granulosa cells, in folliculogenesis? DESIGN: mRNA expression of PEDF and other key factors [Cyp19, anti-Müllerian hormone receptor (AMHR) and vascular endothelial growth factor (VEGF)] in mice follicles was examined in order to typify the expression of PEDF in growing follicles and in human primary granulosa cells (hpGC), and to follow the interplay between PEDF and the other main players in folliculogenesis: FSH and AMH. RESULTS: mRNA expression of PEDF increased through folliculogenesis, although the pattern differed from that of the other examined genes, affecting the follicular angiogenic and oxidative balance. In hpGC, prolonged exposure to FSH stimulated the up-regulation of PEDF mRNA. Furthermore, a negative correlation between AMH and PEDF was observed: AMH stimulation reduced the expression of PEDF mRNA and PEDF stimulation reduced the expression of AMHR mRNA. CONCLUSIONS: Folliculogenesis, an intricate process that requires close dialogue between the oocyte and its supporting granulosa cells, is mediated by various endocrine and paracrine factors. The current findings suggest that PEDF, expressed in granulosa cells, is a pro-folliculogenesis player that interacts with FSH and AMH in the process of follicular growth. However, the mechanism of this process is yet to be determined.


Sujet(s)
Hormone antimullérienne , Protéines de l'oeil , Cellules de la granulosa , Facteurs de croissance nerveuse , Follicule ovarique , Serpines , Serpines/métabolisme , Serpines/génétique , Facteurs de croissance nerveuse/métabolisme , Facteurs de croissance nerveuse/génétique , Femelle , Protéines de l'oeil/métabolisme , Protéines de l'oeil/génétique , Animaux , Cellules de la granulosa/métabolisme , Follicule ovarique/métabolisme , Humains , Souris , Hormone antimullérienne/métabolisme , Hormone folliculostimulante/métabolisme , Hormone folliculostimulante/pharmacologie , Récepteurs peptidiques/métabolisme , Récepteurs peptidiques/génétique , ARN messager/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/génétique , Cellules cultivées
15.
Theriogenology ; 226: 228-235, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38924892

RÉSUMÉ

Although anti-Müllerian hormone (AMH) is involved in the regulation of granulosa cell function in female animals, its role in tissues other than ovarian follicles remains poorly understood. It has also been suggested that cows with high circulating AMH concentrations have increased fertility; however, the mechanism has not been elucidated. This study was conducted to identify the presence of the AMH-signaling system and its target cells in the bovine corpus luteum formed from an ovulated follicle. Immunoblotting revealed that the proteolytically cleaved C-terminal region in AMH (AMHC), a biologically active peptide, was present in trace amounts in the early corpus luteum and significantly increased during the mid to regressed stages. AMHC and cleaved N-terminal region (AMHN) in AMH generate a noncovalent isoform that improves the activity of AMH signaling. An immunohistochemical analysis revealed that AMHC, AMHN, and type II AMH receptor (AMHR2) were localized to luteal cells during the entire estrous cycle. AMH in the corpus luteum seemed to be newly synthesized since AMH expression was detected. These findings suggest that AMH signaling is involved in the regulation of luteal cell function through an autocrine and post-translational processing mechanism. The level of AMHR2 and mRNA expression of AMHR2 and type I AMH receptors (activin-like kinase 2, 3, and 6) were highest in the mid stage. Thus, AMH signaling in the corpus luteum may also be regulated by changes in the receptor levels. Since the transforming growth factor-beta superfamily, to which AMH belongs, is a multifunctional polypeptide growth factor, further studies are needed to evaluate whether AMH signaling has a role in facilitating or inhibiting luteal cell functions.


Sujet(s)
Hormone antimullérienne , Corps jaune , Récepteurs peptidiques , Récepteurs TGF-bêta , Animaux , Femelle , Hormone antimullérienne/métabolisme , Hormone antimullérienne/génétique , Corps jaune/métabolisme , Bovins , Récepteurs peptidiques/métabolisme , Récepteurs peptidiques/génétique , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/génétique , Régulation de l'expression des gènes/physiologie , Cycle oestral/métabolisme , Cycle oestral/physiologie , ARN messager/métabolisme , ARN messager/génétique
16.
Semin Reprod Med ; 42(1): 41-48, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38908381

RÉSUMÉ

Anti-Müllerian hormone (AMH) is an important component within androgen receptor (AR)-regulated pathways governing the hyperandrogenic origin of polycystic ovary syndrome (PCOS). In women with PCOS, granulosa cell AMH overexpression in developing ovarian follicles contributes to elevated circulating AMH levels beginning at birth and continuing in adolescent daughters of PCOS women. A 6 to 7% incidence among PCOS women of gene variants coding for AMH or its receptor, AMHR2, suggests genetic contributions to AMH-related pathogenesis. Discrete gestational AMH administration to pregnant mice induces hypergonadotropic hyperandrogenic, PCOS-like female offspring with high circulating AMH levels that persist over three generations, suggesting epigenetic contributions to PCOS through developmental programming. Moreover, adult-onset, selective hyperactivation of hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH) induces hypergonadotropic hyperandrogenism and PCOS-like traits in female mice. Both gestational and adult AMH inductions of PCOS-like traits are prevented by GnRH antagonist coadministration, implicating luteinizing hormone-dependent ovarian theca cell testosterone (T) action, mediated through the AR in AMH-induced pathogenesis. Interestingly, gestational or peripubertal exogenous T or dihydrotestosterone induction of PCOS-like traits in female mice, rats, sheep, and monkeys fails to elicit ovarian AMH hypersecretion; thus, AMH excess per se may lead to a distinct pathogenic contribution to hyperandrogenic PCOS origins.


Sujet(s)
Hormone antimullérienne , Syndrome des ovaires polykystiques , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/génétique , Hormone antimullérienne/métabolisme , Femelle , Animaux , Humains , Grossesse , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Hyperandrogénie/métabolisme , Hyperandrogénie/génétique , Récepteurs peptidiques/métabolisme , Récepteurs peptidiques/génétique , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/génétique , Souris , Hormone de libération des gonadotrophines/métabolisme , Ovaire/métabolisme , Ovaire/anatomopathologie , Testostérone/sang , Testostérone/métabolisme
17.
Biochim Biophys Acta Gen Subj ; 1868(9): 130660, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38871061

RÉSUMÉ

Caveolin-1 is critical for interacting with the TGF-ß receptor (TGFßR) and EGF receptor (EGFR) signaling, often observed in advanced cancers and tissue fibrosis. However, the mechanism underlying caveolin-1-mediated transactivation of TGFßR and EGFR signaling remains unclear. Therefore, we sought to determine whether caveolin-1 is involved in canonical and non-canonical TGFßR and EGFR signaling transactivation in this study. Methyl-ß-cyclodextrin (MßCD) was used to disrupt the cholesterol-containing membranes domains, and the caveolin-1 scaffolding domain (CSD) peptide was used to mimic the CSD of caveolin-1. Additionally, we transfected the Madin-Darby canine kidney cells with wild-type or phosphorylation-defective caveolin-1. We discovered that tyrosine 14 of caveolin-1 was critical for the negative regulation of TGFßR and EGFR canonical signaling. On the contrary, caveolin-1 inhibited TGF-ß1-induced ERK2 activation independent of tyrosine 14 phosphorylation. Although EGF failed to induce Smad3 phosphorylation in caveolin-1 knockdown cells, it activated Smad3 upon MßCD co-treatment, indicating that caveolin-1 indirectly regulated the non-canonical pathway of EGF. In conclusion, caveolin-1 differentially modulates TGFßR and EGFR signaling. Thus, targeting caveolin-1 is a potential strategy for treating diseases involving TGF-ß1 and EGF signaling.


Sujet(s)
Cavéoline-1 , Récepteurs ErbB , Transduction du signal , Animaux , Chiens , Cavéoline-1/métabolisme , Cavéoline-1/génétique , Cellules rénales canines Madin-Darby , Récepteurs ErbB/métabolisme , Phosphorylation , Facteur de croissance épidermique/métabolisme , Facteur de croissance épidermique/pharmacologie , Facteur de croissance transformant bêta/métabolisme , Récepteurs TGF-bêta/métabolisme , Humains , Facteur de croissance transformant bêta-1/métabolisme
18.
PLoS Comput Biol ; 20(5): e1012072, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38753874

RÉSUMÉ

Cells use signaling pathways to sense and respond to their environments. The transforming growth factor-ß (TGF-ß) pathway produces context-specific responses. Here, we combined modeling and experimental analysis to study the dependence of the output of the TGF-ß pathway on the abundance of signaling molecules in the pathway. We showed that the TGF-ß pathway processes the variation of TGF-ß receptor abundance using Liebig's law of the minimum, meaning that the output-modifying factor is the signaling protein that is most limited, to determine signaling responses across cell types and in single cells. We found that the abundance of either the type I (TGFBR1) or type II (TGFBR2) TGF-ß receptor determined the responses of cancer cell lines, such that the receptor with relatively low abundance dictates the response. Furthermore, nuclear SMAD2 signaling correlated with the abundance of TGF-ß receptor in single cells depending on the relative expression levels of TGFBR1 and TGFBR2. A similar control principle could govern the heterogeneity of signaling responses in other signaling pathways.


Sujet(s)
Transduction du signal , Facteur de croissance transformant bêta , Facteur de croissance transformant bêta/métabolisme , Humains , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Récepteur de type II du facteur de croissance transformant bêta/génétique , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Récepteur de type I du facteur de croissance transformant bêta/génétique , Protéine Smad2/métabolisme , Biologie informatique , Modèles biologiques , Lignée cellulaire tumorale , Protéines Smad/métabolisme , Récepteurs TGF-bêta/métabolisme
19.
Bioorg Med Chem Lett ; 108: 129797, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38759932

RÉSUMÉ

TGF-ß is an immunosuppressive cytokine and plays a key role in progression of cancer by inducing immunosuppression in tumor microenvironment. Therefore, inhibition of TGF-ß signaling pathway may provide a potential therapeutic intervention in treating cancers. Herein, we report the discovery of a series of novel thiazole derivatives as potent inhibitors of ALK5, a serine-threonine kinase which is responsible for TGF-ß signal transduction. Compound 29b was identified as a potent inhibitor of ALK5 with an IC50 value of 3.7 nM with an excellent kinase selectivity.


Sujet(s)
Conception de médicament , Inhibiteurs de protéines kinases , Protein-Serine-Threonine Kinases , Récepteur de type I du facteur de croissance transformant bêta , Récepteurs TGF-bêta , Thiazoles , Thiazoles/composition chimique , Thiazoles/pharmacologie , Thiazoles/synthèse chimique , Récepteur de type I du facteur de croissance transformant bêta/antagonistes et inhibiteurs , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Humains , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Relation structure-activité , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/métabolisme , Récepteurs TGF-bêta/antagonistes et inhibiteurs , Récepteurs TGF-bêta/métabolisme , Structure moléculaire , Relation dose-effet des médicaments
20.
Radiat Res ; 202(1): 1-10, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38772553

RÉSUMÉ

As the number of cancer survivors increases and the risk of accidental radiation exposure rises, there is a pressing need to characterize the delayed effects of radiation exposure and develop medical countermeasures. Radiation has been shown to damage adipose progenitor cells and increase liver fibrosis, such that it predisposes patients to developing metabolic-associated fatty liver disease (MAFLD) and insulin resistance. The risk of developing these conditions is compounded by the global rise of diets rich in carbohydrates and fats. Radiation persistently increases the signaling cascade of transforming growth factor ß (TGFß), leading to heightened fibrosis as characteristic of the delayed effects of radiation exposure. We investigate here a potential radiation medical countermeasure, IPW-5371, a small molecule inhibitor of TGFßRI kinase (ALK5). We found that mice exposed to sub-lethal whole-body irradiation and chronic Western diet consumption but treated with IPW-5371 had a similar body weight, food consumption, and fat mass compared to control mice exposed to radiation. The IPW-5371 treated mice maintained lower fibrosis and fat accumulation in the liver, were more responsive to insulin and had lower circulating triglycerides and better muscle endurance. Future studies are needed to verify the improvement by IPW-5371 on the structure and function of other metabolically active tissues such as adipose and skeletal muscle, but these data demonstrate that IPW-5371 protects liver and whole-body health in rodents exposed to radiation and a Western diet, and there may be promise in using IPW-5371 to prevent the development of MAFLD.


Sujet(s)
Stéatose hépatique , Insulinorésistance , Animaux , Souris , Stéatose hépatique/prévention et contrôle , Stéatose hépatique/anatomopathologie , Mâle , Souris de lignée C57BL , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/antagonistes et inhibiteurs , Régime alimentaire , Irradiation corporelle totale/effets indésirables
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE