Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(6): 107300, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641066

ABSTRACT

Integrin-mediated activation of the profibrotic mediator transforming growth factor-ß1 (TGF-ß1), plays a critical role in idiopathic pulmonary fibrosis (IPF) pathogenesis. Galectin-3 is believed to contribute to the pathological wound healing seen in IPF, although its mechanism of action is not precisely defined. We hypothesized that galectin-3 potentiates TGF-ß1 activation and/or signaling in the lung to promote fibrogenesis. We show that galectin-3 induces TGF-ß1 activation in human lung fibroblasts (HLFs) and specifically that extracellular galectin-3 promotes oleoyl-L-α-lysophosphatidic acid sodium salt-induced integrin-mediated TGF-ß1 activation. Surface plasmon resonance analysis confirmed that galectin-3 binds to αv integrins, αvß1, αvß5, and αvß6, and to the TGFßRII subunit in a glycosylation-dependent manner. This binding is heterogeneous and not a 1:1 binding stoichiometry. Binding interactions were blocked by small molecule inhibitors of galectin-3, which target the carbohydrate recognition domain. Galectin-3 binding to ß1 integrin was validated in vitro by coimmunoprecipitation in HLFs. Proximity ligation assays indicated that galectin-3 and ß1 integrin colocalize closely (≤40 nm) on the cell surface and that colocalization is increased by TGF-ß1 treatment and blocked by galectin-3 inhibitors. In the absence of TGF-ß1 stimulation, colocalization was detectable only in HLFs from IPF patients, suggesting the proteins are inherently more closely associated in the disease state. Galectin-3 inhibitor treatment of precision cut lung slices from IPF patients' reduced Col1a1, TIMP1, and hyaluronan secretion to a similar degree as TGF-ß type I receptor inhibitor. These data suggest that galectin-3 promotes TGF-ß1 signaling and may induce fibrogenesis by interacting directly with components of the TGF-ß1 signaling cascade.


Subject(s)
Fibroblasts , Galectin 3 , Idiopathic Pulmonary Fibrosis , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/metabolism , Galectin 3/metabolism , Galectin 3/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Signal Transduction , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptors, Transforming Growth Factor beta/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Galectins/metabolism , Collagen Type I/metabolism , Cells, Cultured , Blood Proteins
2.
PLoS Comput Biol ; 20(5): e1012072, 2024 May.
Article in English | MEDLINE | ID: mdl-38753874

ABSTRACT

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.


Subject(s)
Signal Transduction , Transforming Growth Factor beta , Transforming Growth Factor beta/metabolism , Humans , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Smad2 Protein/metabolism , Computational Biology , Models, Biological , Cell Line, Tumor , Smad Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism
3.
BMC Biol ; 22(1): 141, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926709

ABSTRACT

BACKGROUND: The Percidae family comprises many fish species of major importance for aquaculture and fisheries. Based on three new chromosome-scale assemblies in Perca fluviatilis, Perca schrenkii, and Sander vitreus along with additional percid fish reference genomes, we provide an evolutionary and comparative genomic analysis of their sex-determination systems. RESULTS: We explored the fate of a duplicated anti-Mullerian hormone receptor type-2 gene (amhr2bY), previously suggested to be the master sex-determining (MSD) gene in P. flavescens. Phylogenetically related and structurally similar amhr2 duplicates (amhr2b) were found in P. schrenkii and Sander lucioperca, potentially dating this duplication event to their last common ancestor around 19-27 Mya. In P. fluviatilis and S. vitreus, this amhr2b duplicate has been likely lost while it was subject to amplification in S. lucioperca. Analyses of the amhr2b locus in P. schrenkii suggest that this duplication could be also male-specific as it is in P. flavescens. In P. fluviatilis, a relatively small (100 kb) non-recombinant sex-determining region (SDR) was characterized on chromosome 18 using population-genomics approaches. This SDR is characterized by many male-specific single-nucleotide variations (SNVs) and no large duplication/insertion event, suggesting that P. fluviatilis has a male heterogametic sex-determination system (XX/XY), generated by allelic diversification. This SDR contains six annotated genes, including three (c18h1orf198, hsdl1, tbc1d32) with higher expression in the testis than in the ovary. CONCLUSIONS: Together, our results provide a new example of the highly dynamic sex chromosome turnover in teleosts and provide new genomic resources for Percidae, including sex-genotyping tools for all three known Perca species.


Subject(s)
Evolution, Molecular , Sex Determination Processes , Animals , Sex Determination Processes/genetics , Male , Female , Perches/genetics , Phylogeny , Receptors, Peptide/genetics , Genome , Receptors, Transforming Growth Factor beta
4.
Genesis ; 62(4): e23614, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39139086

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Developmental , Larva , Paracentrotus , Animals , Larva/growth & development , Larva/metabolism , Larva/genetics , Paracentrotus/genetics , Paracentrotus/metabolism , Paracentrotus/embryology , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Osteogenesis/genetics , Gene Regulatory Networks , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
Mol Pharmacol ; 105(4): 286-300, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38278554

ABSTRACT

Prodigiosin (PG) is a naturally occurring polypyrrole red pigment produced by numerous microorganisms including some Serratia and Streptomyces strains. PG has exhibited promising anticancer activity; however, the molecular mechanisms of action of PG on malignant cells remain ambiguous. Transforming growth factor-ß (TGF-ß) is a multifunctional cytokine that governs a wide array of cellular processes in development and tissue homeostasis. Malfunctions of TGF-ß signaling are associated with numerous human cancers. Emerging evidence underscores the significance of internalized TGF-ß receptors and their intracellular trafficking in initiating signaling cascades. In this study, we identified PG as a potent inhibitor of the TGF-ß pathway. PG blocked TGF-ß signaling by targeting multiple sites of this pathway, including facilitating the sequestering of TGF-ß receptors in the cytoplasm by impeding the recycling of type II TGF-ß receptors to the cell surface. Additionally, PG prompts a reduction in the abundance of receptors on the cell surface through the disruption of the receptor glycosylation. In human Caucasian lung carcinoma cells and human hepatocellular cancer cell line cells, nanomolar concentrations of PG substantially diminish TGF-ß-triggered phosphorylation of Smad2 protein. This attenuation is further reflected in the suppression of downstream target gene expression, including those encoding fibronectin, plasminogen activator inhibitor-1, and N-cadherin. SIGNIFICANCE STATEMENT: Prodigiosin (PG) emerges from this study as a potent TGF-ß pathway inhibitor, disrupting receptor trafficking and glycosylation and reducing TGF-ß signaling and downstream gene expression. These findings not only shed light on PG's potential therapeutic role but also present a captivating avenue towards future anti-TGF-ß strategies.


Subject(s)
Protein Serine-Threonine Kinases , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Protein Serine-Threonine Kinases/metabolism , Prodigiosin/pharmacology , Prodigiosin/metabolism , Polymers/metabolism , Pyrroles , Receptors, Transforming Growth Factor beta/metabolism , Phosphorylation , Epithelial Cells/metabolism , Transforming Growth Factor beta1 , Smad2 Protein/metabolism
6.
J Recept Signal Transduct Res ; 44(1): 27-34, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38660706

ABSTRACT

Diabetic kidney disease (DKD) is one of the most serious complications of diabetes and has become the leading cause of end-stage kidney disease, causing serious health damage and a huge economic burden. Tubulointerstitial fibrosis play important role in the development of DKD. Itaconate, a macrophage-specific metabolite, has been reported to have anti-oxidant, anti-inflammatory effects. However, it is unknown whether it perform anti-fibrotic effect in renal tubular epithelial cells. In this current study, we observed that in human renal tubular epithelial cells (HK2), high glucose induced an increase in transforming growth factor ß (TGF-ß) production, and upregulated the expressions of fibronectin and collagen I through the TGF-ß receptor as verified by administration of TGF-ß receptor blocker LY2109761. Treatment with 4-octyl itaconate (4-OI), a derivant of itaconic acid, reduced the TGF-ß production induced by high glucose and inhibited the pro-fibrotic effect of TGF-ß in a dose-dependent manner. In addition, we found that 4-OI exerted its anti-fibrotic effect by inhibiting the excessive production of ROS induced by high glucose and TGF-ß. In summary, 4-OI could ameliorate high glucose-induced pro-fibrotic effect in HK2 cell, and blocking the expression of TGF-ß and reducing the excessive ROS production may be involved in its anti-fibrotic effect.


Subject(s)
Glucose , Kidney Tubules , Signal Transduction , Succinates , Humans , Cell Line , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Diabetic Nephropathies/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/metabolism , Fibronectins/metabolism , Fibronectins/genetics , Fibrosis/drug therapy , Glucose/metabolism , Kidney Tubules/pathology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Pyrazoles , Pyrroles/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Succinates/pharmacology , Transforming Growth Factor beta/metabolism
7.
Exp Dermatol ; 33(8): e15121, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39081004

ABSTRACT

Dermatofibrosarcoma protuberans (DFSP) is a rare cutaneous sarcoma characterized by the COL1A1-PDGFB fusion gene. This study utilized single-cell RNA sequencing to dissect the cellular and molecular landscape of primary DFSP. Distinct DFSP cell clusters, exhibiting fibroblast-like traits, revealed variations in pathways associated with proliferation, inflammation and metabolism. Differential gene expression analysis during the differentiation from tumour stem cells to DFSP cells unveiled SMOC2, DCN and TGFBR3 as potential regulators of tumour invasion and immune infiltration through VEGF/TGF-ß signalling modulation. Cellular communication analysis highlighted interactions within DFSP cell clusters and with endothelial cells, implicating molecules such as NAMPT, ANGPT2 and PTN in pathogenesis and treatment resistance. These findings offer insights into DFSP intratumour heterogeneity, elucidate molecular mechanisms underlying tumour behaviour, and suggest potential therapeutic targets.


Subject(s)
Dermatofibrosarcoma , Single-Cell Analysis , Skin Neoplasms , Dermatofibrosarcoma/genetics , Dermatofibrosarcoma/pathology , Dermatofibrosarcoma/metabolism , Humans , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Sequence Analysis, RNA , Cell Communication/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Cell Differentiation , RNA-Seq , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Proteoglycans , Receptors, Transforming Growth Factor beta
8.
BMC Cancer ; 24(1): 444, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600507

ABSTRACT

BACKGROUND: Transforming growth factor-ß (TGF-ß) is a cytokine with multiple functions, including cell growth regulation, extracellular matrix production, angiogenesis homeostasis adjustment and et al. TGF-ß pathway activation promotes tumor metastasis/progression and mediates epithelial-mesenchymal transmission suppressing immunosurveillance in advanced tumors. GFH018, a small molecule inhibitor blocking TGF-ß signal transduction, inhibits the progression and/or metastasis of advanced cancers. This first-in-human study evaluated the safety, tolerability, pharmacokinetics (PK), and efficacy of GFH018 monotherapy in patients with advanced solid tumors. METHODS: This phase I, open-label, multicenter study used a modified 3+3 dose escalation and expansion design. Adult patients with advanced solid tumors failing the standard of care were enrolled. Starting at 5 mg, eight dose levels up to 85 mg were evaluated. Patients received GFH018 BID (14d-on/14d-off) starting on the 4th day after a single dose on cycle 1, day 1. Subsequent cycles were defined as 28 days. The study also explored the safety of 85 mg BID 7d-on/7d-off. Adverse events were graded using NCI criteria for adverse events (NCI-CTCAE v5.0). PK was analyzed using a noncompartmental method. Efficacy was evaluated using RECIST 1.1. Blood samples were collected for biomarker analysis. RESULTS: Fifty patients were enrolled and received at least one dose of GFH018. No dose-limiting toxicity occurred, and the maximum tolerated dose was not reached. Forty-three patients (86.0%) had at least one treatment-related adverse event (TRAE), and three patients (6.0%) had ≥ G3 TRAEs. The most common TRAEs (any grade/grade ≥3) were AST increased (18%/0%), proteinuria (14%/2%), anemia (14%/2%), and ALT increased (12%/0%). No significant cardiotoxicity or bleeding was observed. GFH018 PK was linear and dose-independent, with a mean half-life of 2.25-8.60 h from 5 - 85 mg. Nine patients (18.0%) achieved stable disease, and one patient with thymic carcinoma achieved tumor shrinkage, with the maximum target lesion decreased by 18.4%. Serum TGF-ß1 levels were not associated with clinical responses. The comprehensive recommended dose for Phase II was defined as 85 mg BID 14d-on/14d-off. CONCLUSIONS: GFH018 monotherapy presented a favorable safety profile without cardiac toxicity or bleeding. Modest efficacy warrants further studies, including combination strategies. TRIAL REGISTRATION: ClinicalTrial. gov ( https://www. CLINICALTRIALS: gov/ ), NCT05051241. Registered on 2021-09-02.


Subject(s)
Neoplasms , Receptors, Transforming Growth Factor beta , Adult , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Response Evaluation Criteria in Solid Tumors , Transforming Growth Factor beta , Receptors, Transforming Growth Factor beta/antagonists & inhibitors
9.
Cell Commun Signal ; 22(1): 128, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38360757

ABSTRACT

In pathologies including cancer, aberrant Transforming Growth Factor-ß (TGF-ß) signaling exerts profound tumor intrinsic and extrinsic consequences. Intense clinical endeavors are underway to target this pathway. Central to the success of these interventions is pinpointing factors that decisively modulate the TGF-ß responses. Betaglycan/type III TGF-ß receptor (TßRIII), is an established co-receptor for the TGF-ß superfamily known to bind directly to TGF-ßs 1-3 and inhibin A/B. Betaglycan can be membrane-bound and also undergo ectodomain cleavage to produce soluble-betaglycan that can sequester its ligands. Its extracellular domain undergoes heparan sulfate and chondroitin sulfate glycosaminoglycan modifications, transforming betaglycan into a proteoglycan. We report the unexpected discovery that the heparan sulfate glycosaminoglycan chains on betaglycan are critical for the ectodomain shedding. In the absence of such glycosaminoglycan chains betaglycan is not shed, a feature indispensable for the ability of betaglycan to suppress TGF-ß signaling and the cells' responses to exogenous TGF-ß ligands. Using unbiased transcriptomics, we identified TIMP3 as a key inhibitor of betaglycan shedding thereby influencing TGF-ß signaling. Our results bear significant clinical relevance as modified betaglycan is present in the ascites of patients with ovarian cancer and can serve as a marker for predicting patient outcomes and TGF-ß signaling responses. These studies are the first to demonstrate a unique reliance on the glycosaminoglycan chains of betaglycan for shedding and influence on TGF-ß signaling responses. Dysregulated shedding of TGF-ß receptors plays a vital role in determining the response and availability of TGF-ßs', which is crucial for prognostic predictions and understanding of TGF-ß signaling dynamics.


Subject(s)
Glycosaminoglycans , Ovarian Neoplasms , Humans , Female , Glycosaminoglycans/metabolism , Transforming Growth Factor beta/metabolism , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Heparitin Sulfate/metabolism
10.
Reprod Biomed Online ; 49(2): 103981, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870625

ABSTRACT

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.


Subject(s)
Anti-Mullerian Hormone , Eye Proteins , Granulosa Cells , Nerve Growth Factors , Ovarian Follicle , Serpins , Serpins/metabolism , Serpins/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Female , Eye Proteins/metabolism , Eye Proteins/genetics , Animals , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Humans , Mice , Anti-Mullerian Hormone/metabolism , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone/pharmacology , Receptors, Peptide/metabolism , Receptors, Peptide/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/genetics , Cells, Cultured
11.
Wound Repair Regen ; 32(3): 234-245, 2024.
Article in English | MEDLINE | ID: mdl-38459905

ABSTRACT

Cells integrate many mechanical and chemical cues to drive cell signalling responses. Because of the complex nature and interdependency of alterations in extracellular matrix (ECM) composition, ligand density, mechanics, and cellular responses it is difficult to tease out individual and combinatorial contributions of these various factors in driving cell behavior in homeostasis and disease. Tuning of material viscous and elastic properties, and ligand densities, in combinatorial fashions would enhance our understanding of how cells process complex signals. For example, it is known that increased ECM mechanics and transforming growth factor beta (TGF-ß) receptor (TGF-ß-R) spacing/clustering independently drive TGF-ß signalling and associated myofibroblastic differentiation. However, it remains unknown how these inputs orthogonally contribute to cellular outcomes. Here, we describe the development of a novel material platform that combines microgel thin films with controllable viscoelastic properties and DNA origami to probe how viscoelastic properties and nanoscale spacing of TGF-ß-Rs contribute to TGF-ß signalling and myofibroblastic differentiation. We found that highly viscous materials with non-fixed TGF-ß-R spacing promoted increased TGF-ß signalling and myofibroblastic differentiation. This is likely due to the ability of cells to better cluster receptors on these surfaces. These results provide insight into the contribution of substrate properties and receptor localisation on downstream signalling. Future studies allow for exploration into other receptor-mediated processes.


Subject(s)
Biocompatible Materials , Cell Differentiation , Extracellular Matrix , Myofibroblasts , Signal Transduction , Transforming Growth Factor beta , Biocompatible Materials/pharmacology , Myofibroblasts/metabolism , Myofibroblasts/physiology , Transforming Growth Factor beta/metabolism , Extracellular Matrix/metabolism , Humans , Receptors, Transforming Growth Factor beta/metabolism , Cells, Cultured , Tissue Engineering/methods , Viscosity
12.
Bioorg Med Chem Lett ; 108: 129797, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38759932

ABSTRACT

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.


Subject(s)
Drug Design , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Thiazoles , Thiazoles/chemistry , Thiazoles/pharmacology , Thiazoles/chemical synthesis , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Molecular Structure , Dose-Response Relationship, Drug
13.
Bioorg Chem ; 146: 107274, 2024 May.
Article in English | MEDLINE | ID: mdl-38503026

ABSTRACT

The inhibition of transforming growth factor-ß1 (TGF-ß1) signaling by targeting TGF-ß receptor 1 (TßR1) has been considered as an ideal approach for the prevention of pancreatic cancer metastasis. Utilizing a pharmacophore model for TßR1 inhibitors, candidate compounds with the potential TßR1 binding ability were screened from the U.S. Food and Drug Administration (FDA) database, and riboflavin (RF) with a highest fit value was chosen to investigate its binding ability to TßR1 and effect on TGF-ß1 signaling in pancreatic cancer cells. Molecular docking and cellular thermal shift assay (CETSA) proved that RF at pharmacological concentrations could directly bind to TßR1. Further studies showed that pharmacological concentrations of RF in vitro could block TGF-ß1 signaling, suppress the migration and invasion, and prevent epithelial-mesenchymal transition (EMT) process of pancreatic cancer cells in the absence or presence of TGF-ß1 stimulation, indicating that RF presented anti-metastatic effect in pancreatic cancer cells. Knockdown of TßR1 could significantly attenuate the effects of RF on the migration and EMT process in pancreatic cancer cells, further confirming that the anti-metastatic effect of RF was achieved by blocking TGF-ß1 signaling after binding to TßR1. Moreover, in a mouse model of pancreatic cancer metastasis, it was certified that RF administration could block lung and liver metastases, TGF-ß1 signaling and EMT process of pancreatic cancer in vivo. In summary, our findings showed that RF could block TGF-ß1 signaling by directly binding to TßR1, thereby suppressing the metastasis of pancreatic cancer cells by inhibiting EMT process both in vitro and in vivo.


Subject(s)
Pancreatic Neoplasms , Transforming Growth Factor beta1 , Animals , Mice , Transforming Growth Factor beta1/metabolism , Molecular Docking Simulation , Cell Line, Tumor , Neoplasm Invasiveness/prevention & control , Pancreatic Neoplasms/drug therapy , Receptors, Transforming Growth Factor beta , Epithelial-Mesenchymal Transition
14.
Acta Pharmacol Sin ; 45(7): 1337-1348, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38351317

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling is initiated by activation of transmembrane TGF-ß receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-ß signaling pathways leads to pathological conditions. TGF-ß signaling is regulated at different levels along the pathways and begins with the liberation of TGF-ß ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-ß isoforms, enabling precise control of TGF-ß signals. In addition, the cell surface compartments used to release active TGF-ß are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.


Subject(s)
Receptors, Transforming Growth Factor beta , Signal Transduction , Transforming Growth Factor beta , Humans , Receptors, Transforming Growth Factor beta/metabolism , Ligands , Animals , Transforming Growth Factor beta/metabolism
15.
Reprod Domest Anim ; 59(6): e14635, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38837470

ABSTRACT

A 1-year-old European shorthair male cat with a normally developed penis was subjected to genetic, endocrinological and histological studies due to unilateral cryptorchidism. The blood testosterone level was typical for males, while the level of anti-Mullerian hormone (AMH) was very low. Surgical removal of internal reproductive organs was followed by a histological study, which revealed inactive testicles with neoplastic changes and derivatives of Mullerian ducts. Cytogenetic analysis showed a normal XY sex chromosome complement and molecular analysis confirmed the presence of Y-linked genes (SRY and ZFY). Although the level of AMH was low, two normal copies of the AMH gene were found using droplet digital PCR (ddPCR). Analysis of the coding sequences of two candidate genes (AMH and AMHR2) for persistent Mullerian duct syndrome (PMDS) in the affected cat and in control male cats (n = 24) was performed using the Sanger sequencing method. In the affected cat, homozygosity was found for three novel missense variants in Exon 1 (one SNP) and Exon 5 (two SNPs) of AMH, but the same homozygous genotypes were also observed in one and two control cats, respectively, whose sex development was not examined. Three known synonymous variants with homozygous status were found in AMHR2. We conclude that the DNA variants identified in AMH and AMHR2 are not responsible for PMDS in the affected cat.


Subject(s)
Anti-Mullerian Hormone , Cat Diseases , Receptors, Peptide , Receptors, Transforming Growth Factor beta , Animals , Cats , Male , Anti-Mullerian Hormone/genetics , Cat Diseases/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Cryptorchidism/genetics , Cryptorchidism/veterinary , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/veterinary , Mutation , Mutation, Missense
16.
Arch Pharm (Weinheim) ; 357(8): e2400131, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38678538

ABSTRACT

Three series of N-{[4-([1,2,4]triazolo[1,5-α]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-yl]methyl}acetamides (14a-d, 15a-n, and 16a-f) were synthesized and evaluated for activin receptor-like kinase 5 (ALK5) inhibitory activities in an enzymatic assay. The target compounds showed high ALK5 inhibitory activity and selectivity. The half maximal inhibitory concentration (IC50) for phosphorylation of ALK5 of 16f (9.1 nM), the most potent compound, was 2.7 times that of the clinical candidate EW-7197 (vactosertib) and 14 times that of the clinical candidate LY-2157299. The selectivity index of 16f against p38α mitogen-activated protein kinase was >109, which was much higher than that of positive controls (EW-7197: >41, and LY-2157299: 4). Furthermore, a molecular docking study provided the interaction modes between the target compounds and ALK5. Compounds 14c, 14d, and 16f effectively inhibited the protein expression of α-smooth muscle actin (α-SMA), collagen I, and tissue inhibitor of metalloproteinase 1 (TIMP-1)/matrix metalloproteinase 13 (MMP-13) in transforming growth factor-ß-induced human umbilical vein endothelial cells. Compounds 14c and 16f showed especially high activity at low concentrations, which suggests that these compounds could inhibit myocardial cell fibrosis. Compounds 14c, 14d, and 16f are potential preclinical candidates for the treatment of cardiac fibrosis.


Subject(s)
Fibrosis , Imidazoles , Molecular Docking Simulation , Receptor, Transforming Growth Factor-beta Type I , Humans , Imidazoles/pharmacology , Imidazoles/chemical synthesis , Imidazoles/chemistry , Structure-Activity Relationship , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Fibrosis/drug therapy , Molecular Structure , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Antifibrotic Agents/pharmacology , Antifibrotic Agents/chemical synthesis , Antifibrotic Agents/chemistry , Amides/pharmacology , Amides/chemical synthesis , Amides/chemistry , Dose-Response Relationship, Drug , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism
17.
Bull Exp Biol Med ; 176(5): 603-606, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38730107

ABSTRACT

Polymorphism of genes of transforming growth factor TGFB and its receptors (TGFBRI, TGFBRII, and TGFBRIIII) in patients with primary open-angle glaucoma was analyzed. The frequency of the TGFBRII CC genotype in patients is increased relative to the control group (OR=6.10, p=0.0028). Heterozygosity in this polymorphic position is reduced (OR=0.18, p=0.0052). As the effects of TGF-ß is mediated through its receptors, we analyzed complex of polymorphic variants of the studied loci in the genome of patients. Two protective complexes consisting only of receptor genes were identified: TGFBRI TT:TGFBRII CG (OR=0.10, p=0.02) and TGFBRII CG:TGFBRIII CG (OR=0.09, p=0.01). The study showed an association of TGFBRII polymorphism with primary open-angle glaucoma and the need to study functionally related genes in the development of the disease, which should contribute to its early diagnosis and prevention.


Subject(s)
Glaucoma, Open-Angle , Humans , Glaucoma, Open-Angle/genetics , Female , Male , Middle Aged , Siberia , Aged , Polymorphism, Single Nucleotide/genetics , Genetic Predisposition to Disease/genetics , Receptors, Transforming Growth Factor beta/genetics , Gene Frequency/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Case-Control Studies , Genotype , Transforming Growth Factor beta/genetics , Receptor, Transforming Growth Factor-beta Type I/genetics , Polymorphism, Genetic/genetics
18.
Radiat Res ; 202(1): 1-10, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38772553

ABSTRACT

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.


Subject(s)
Fatty Liver , Insulin Resistance , Animals , Mice , Fatty Liver/prevention & control , Fatty Liver/pathology , Male , Mice, Inbred C57BL , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Diet , Whole-Body Irradiation/adverse effects
19.
Expert Opin Ther Pat ; 34(3): 99-126, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38648107

ABSTRACT

INTRODUCTION: The TGF-ß signaling pathway is a complex network that plays a crucial role in regulating essential biological functions and is implicated in the onset and progression of multiple diseases. This review highlights the recent advancements in developing inhibitors targeting the TGF-ß signaling pathway and their potential therapeutic applications in various diseases. AREA COVERED: The review discusses patents on active molecules related to the TGF-ß signaling pathway, focusing on three strategies: TGF-ß activity inhibition, blocking TGF-ß receptor binding, and disruption of the signaling pathway using small molecule inhibitors. Combination therapies and the development of fusion proteins targeting multiple pathways are also explored. The literature search was conducted using the Cortellis Drug Discovery Intelligence database, covering patents from 2021 onwards. EXPERT OPINION: The development of drugs targeting the TGF-ß signaling pathway has made significant progress in recent years. However, addressing challenges such as specificity, systemic toxicity, and patient selection is crucial for their successful clinical application. Targeting the TGF-ß signaling pathway holds promise as a promising approach for the treatment of various diseases.


Subject(s)
Drug Development , Molecular Targeted Therapy , Patents as Topic , Receptors, Transforming Growth Factor beta , Signal Transduction , Transforming Growth Factor beta , Humans , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Drug Discovery
20.
Eur J Med Chem ; 269: 116311, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38508118

ABSTRACT

Four series of imidazoles (15a-g, 20c, and 20d) and thiazoles (18a-g, 22a, and 22b) possessing various amino acids were synthesized and evaluated for activin receptor-like kinase 5 (ALK5) inhibitory activities in an enzymatic assay. Among them, compounds 15g and 18c showed the highest inhibitory activity against ALK5, with IC50 values of 0.017 and 0.025 µM, respectively. Compounds 15g and 18c efficiently inhibited extracellular matrix (ECM) deposition in TGF-ß-induced hepatic stellate cells (HSCs), and eventually suppressed HSC activation. Moreover, compound 15g showed a good pharmacokinetic (PK) profile with a favorable half-life (t1/2 = 9.14 h). The results indicated that these compounds exhibited activity targeting ALK5 and may have potential in the treatment of liver fibrosis; thus they are worthy of further study.


Subject(s)
Amino Acids , Thiazoles , Humans , Thiazoles/pharmacology , Amino Acids/pharmacology , Liver Cirrhosis/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Imidazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL