Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.375
Filtrer
1.
BMC Vet Res ; 20(1): 344, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097704

RÉSUMÉ

Porcine reproductive and respiratory syndrome virus (PRRSV) induces a poor innate immune response following infection. This study evaluates the effects of transforming growth factor beta 1 (TGFß1) up-regulated by PRRSV on gene expressions of co-stimulatory molecules, type I interferon (IFN), type I IFN-regulated genes (IRGs), pattern recognition receptors, and pro-inflammatory cytokines in PRRSV-inoculated monocyte-derived macrophages (MDMs). Phosphorothioate-modified antisense oligodeoxynucleotides (AS ODNs) specific to various regions of porcine TGFß1 mRNA were synthesized, and those specific to the AUG region efficiently knockdown TGFß1 mRNA expression and protein translation. Transfection of TGFßAS ODNs in MDMs inoculated with either classical PRRSV-2 (cPRRSV-2) or highly pathogenic PRRSV-2 (HP-PRRSV-2) significantly reduced TGFß1 mRNA expression and significantly increased mRNA expressions of CD80, CD86, IFNß, IRGs (i.e. IFN regulatory factor 3 (IRF3), IRF7, myxovirus resistance 1, osteopontin, and stimulator of IFN genes), Toll-like receptor 3, and tumor necrosis factor-alpha. Transfection of TGFßAS ODNs in MDMs inoculated with HP-PRRSV-2 also significantly increased mRNA expressions of IFNα, IFNγ, and 2'-5'-oligoadenylate synthetase 1. The quantity of PRRSV-2 RNA copy numbers was significantly reduced in MDMs transfected with TGFßAS ODNs as compared to untransfected MDMs. Recombinant porcine TGFß1 (rTGFß1) and recombinant porcine IFNα (rIFNα) sustained and reduced the yields of PRRSV-2 RNA copy numbers in PRRSV-2 inoculated MDMs, respectively. These findings demonstrate a strategy of PRRSV for innate immune suppression via an induction of TGFß expression. These findings also suggest TGFß as a potential parameter that future PRRSV vaccine and vaccine adjuvant candidates should take into consideration.


Sujet(s)
Cytokines , Interféron de type I , Macrophages , Virus du syndrome respiratoire et reproducteur porcin , Animaux , Virus du syndrome respiratoire et reproducteur porcin/physiologie , Suidae , Interféron de type I/métabolisme , Cytokines/génétique , Cytokines/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/virologie , Macrophages/immunologie , Macrophages/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Syndrome dysgénésique et respiratoire porcin/immunologie , Syndrome dysgénésique et respiratoire porcin/virologie , Techniques de knock-down de gènes , Immunité innée
2.
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
3.
J Clin Invest ; 134(16)2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39145443

RÉSUMÉ

The phenotypic switch of vascular smooth cells (VSMCs) from a contractile to a synthetic state is associated with the development and progression of aortic aneurysm (AA). However, the mechanism underlying this process remains unclear. In this issue of the JCI, Song et al. identified SLC44A2 as a regulator of the phenotypic switch in VSMCs. Inhibition of SLC44A2 facilitated the switch to the synthetic state, contributing to the development of AA. Mechanistically, SLC44A2 interacted with NRP1 and ITGB3 to activate the TGF-ß/SMAD signaling pathway, resulting in VSMCs with a contractile phenotype. Furthermore, VSMC-specific SLC44A2 overexpression by genetic or pharmacological manipulation reduced AA in mouse models. These findings suggest the potential of targeting the SLC44A2 signaling pathway for AA prevention and treatment.


Sujet(s)
Anévrysme de l'aorte , Muscles lisses vasculaires , Myocytes du muscle lisse , Transduction du signal , Animaux , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Anévrysme de l'aorte/métabolisme , Anévrysme de l'aorte/anatomopathologie , Anévrysme de l'aorte/génétique , Souris , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Humains , Phénotype , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Intégrine bêta3/métabolisme , Intégrine bêta3/génétique , Protéines de transport membranaire/métabolisme , Protéines de transport membranaire/génétique , Neuropiline 1/métabolisme , Neuropiline 1/génétique
4.
Sci Rep ; 14(1): 17884, 2024 08 02.
Article de Anglais | MEDLINE | ID: mdl-39095553

RÉSUMÉ

Colorectal cancer (CRC) is the third most common cancer in the United States. Recent epidemiological evidence demonstrates an increasing incidence of young-onset CRC cases, defined as CRC cases in individuals 50 years old or younger. Studies have established that alterations in both the WNT and TGF-Beta signaling pathways have contributed to CRC development. While this is well understood, the comprehensive analysis of WNT and TGF-Beta pathway alterations in young-onset CRC cases has yet to be investigated. Here, we conducted a comprehensive bioinformatics analysis of mutations associated with each of the WNT and TGF-Beta signaling pathways according to age (≤ 50 years old versus > 50 years old) utilizing published genomic data from the cBioPortal. Chi-square results demonstrated no significant difference in WNT alterations between young-onset CRC and those > 50 years old. However, across all age groups, WNT alterations were frequently found in rectal cancers. We also found that WNT alterations were associated with better outcomes. The mutations associated with TGF-beta were observed at a higher rate in older CRC patients when compared to those ≤ 50 years old. Additionally, these mutations were found more frequently in colon primaries.


Sujet(s)
Âge de début , Tumeurs colorectales , Mutation , Facteur de croissance transformant bêta , Voie de signalisation Wnt , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Adulte d'âge moyen , Voie de signalisation Wnt/génétique , Mâle , Adulte , Femelle , Sujet âgé , Biologie informatique/méthodes , Protéines de type Wingless/génétique , Protéines de type Wingless/métabolisme
5.
Int J Immunopathol Pharmacol ; 38: 3946320241276899, 2024.
Article de Anglais | MEDLINE | ID: mdl-39162714

RÉSUMÉ

BACKGROUND: Induced Pluripotent Stem Cells (IPSCs) represent an innovative strategy for addressing challenging diseases, including various rheumatologic conditions. Aside from their regenerative capacities, some studies have shown the potential of these cells in the modulation of inflammatory responses. The underlying mechanisms by which they exert their effects have yet to be fully comprehended. Therefore, we aimed to explore the gene expression linked to the IGF pathway as well as IL-10 and TGF-ß, which are known to exert immunomodulatory effects. METHODS: A C57/Bl6 pregnant mouse was used for obtaining mouse embryonic fibroblasts (MEFs), then the IPSCs were induced using lentiviral vectors expressing the pluripotency genes (OCT4, SOX2, KLF1, and c-MYC). Cells were cultured for 72 h in DMEM high glucose plus leukemia inhibitory factor; Evaluating the gene expression was conducted using specific primers for Igf1, Igf2, Igfbp3, Igfbp4, Irs1, Il-10, and Tgf-ß genes, as well as SYBR green qPCR master mix. The data were analyzed using the 2-ΔΔCT method and were compared by employing the t test; the results were plotted using GraphPad PRISM software. MEFs were utilized as controls. RESULTS: Gene expression analyses revealed that Igf-1, Igf-bp3, Igf-bp4, and Il-10 were significantly overexpressed (p ≤ .01), while Igf-2 and Tgf-b genes were significantly downregulated in the lysates from IPSCs in comparison with the control MEFs. The Irs1 gene expression was not altered significantly. CONCLUSION: IPSCs are potentially capable of modulating inflammatory responses through the expression of various anti-inflammatory mediators from the IGF signaling, as well as IL-10. This discovery uncovers a previously unknown dimension of IPSCs' therapeutic effects, potentially leading to more advanced in vivo research and subsequent clinical trials.


Sujet(s)
Cellules souches pluripotentes induites , Interleukine-10 , Souris de lignée C57BL , Animaux , Interleukine-10/génétique , Interleukine-10/métabolisme , Cellules souches pluripotentes induites/métabolisme , Souris , Femelle , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Cellules cultivées , Fibroblastes/métabolisme , Grossesse , Immunomodulation/génétique
6.
Nat Genet ; 56(8): 1725-1736, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38951642

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with poor prognosis and limited treatment options. Efforts to identify effective treatments are thwarted by limited understanding of IPF pathogenesis and poor translatability of available preclinical models. Here we generated spatially resolved transcriptome maps of human IPF (n = 4) and bleomycin-induced mouse pulmonary fibrosis (n = 6) to address these limitations. We uncovered distinct fibrotic niches in the IPF lung, characterized by aberrant alveolar epithelial cells in a microenvironment dominated by transforming growth factor beta signaling alongside predicted regulators, such as TP53 and APOE. We also identified a clear divergence between the arrested alveolar regeneration in the IPF fibrotic niches and the active tissue repair in the acutely fibrotic mouse lung. Our study offers in-depth insights into the IPF transcriptional landscape and proposes alveolar regeneration as a promising therapeutic strategy for IPF.


Sujet(s)
Bléomycine , Fibrose pulmonaire idiopathique , Transcriptome , Animaux , Humains , Souris , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Bléomycine/toxicité , Modèles animaux de maladie humaine , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Poumon/métabolisme , Poumon/anatomopathologie , Souris de lignée C57BL , Transduction du signal/génétique , Mâle , Analyse de profil d'expression de gènes , Pneumocytes/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Régénération/génétique , Apolipoprotéines E/génétique
7.
EBioMedicine ; 106: 105228, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39013324

RÉSUMÉ

BACKGROUND: It is uncertain which biological features underpin the response of rectal cancer (RC) to radiotherapy. No biomarker is currently in clinical use to select patients for treatment modifications. METHODS: We identified two cohorts of patients (total N = 249) with RC treated with neoadjuvant radiotherapy (45Gy/25) plus fluoropyrimidine. This discovery set included 57 cases with pathological complete response (pCR) to chemoradiotherapy (23%). Pre-treatment cancer biopsies were assessed using transcriptome-wide mRNA expression and targeted DNA sequencing for copy number and driver mutations. Biological candidate and machine learning (ML) approaches were used to identify predictors of pCR to radiotherapy independent of tumour stage. Findings were assessed in 107 cases from an independent validation set (GSE87211). FINDINGS: Three gene expression sets showed significant independent associations with pCR: Fibroblast-TGFß Response Signature (F-TBRS) with radioresistance; and cytotoxic lymphocyte (CL) expression signature and consensus molecular subtype CMS1 with radiosensitivity. These associations were replicated in the validation cohort. In parallel, a gradient boosting machine model comprising the expression of 33 genes generated in the discovery cohort showed high performance in GSE87211 with 90% sensitivity, 86% specificity. Biological and ML signatures indicated similar mechanisms underlying radiation response, and showed better AUC and p-values than published transcriptomic signatures of radiation response in RC. INTERPRETATION: RCs responding completely to chemoradiotherapy (CRT) have biological characteristics of immune response and absence of immune inhibitory TGFß signalling. These tumours may be identified with a potential biomarker based on a 33 gene expression signature. This could help select patients likely to respond to treatment with a primary radiotherapy approach as for anal cancer. Conversely, those with predicted radioresistance may be candidates for clinical trials evaluating addition of immune-oncology agents and stromal TGFß signalling inhibition. FUNDING: The Stratification in Colorectal Cancer Consortium (S:CORT) was funded by the Medical Research Council and Cancer Research UK (MR/M016587/1).


Sujet(s)
Apprentissage machine , Tumeurs du rectum , Facteur de croissance transformant bêta , Humains , Tumeurs du rectum/génétique , Tumeurs du rectum/radiothérapie , Tumeurs du rectum/anatomopathologie , Tumeurs du rectum/thérapie , Tumeurs du rectum/métabolisme , Tumeurs du rectum/immunologie , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Analyse de profil d'expression de gènes , Transcriptome , Marqueurs biologiques tumoraux/génétique , Résultat thérapeutique , Régulation de l'expression des gènes tumoraux , Pronostic , Adulte
8.
Mol Biol Rep ; 51(1): 831, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39037563

RÉSUMÉ

BACKGROUND: While many genes linked to colorectal cancer (CRC) contribute to cancer development, a thorough investigation is needed to explore crucial hub genes yet to be fully studied. A pivotal pathway in CRC is transforming growth factor-beta (TGF-ß). This study aimed to assess SMAD2 and SMAD4 gene expression from this pathway. METHODS AND RESULTS: Counted data from the Cancer Genome Atlas (TCGA) were examined, comparing 483 tumor and 41 normal samples. Using clinical data, genes impacting overall survival (OS) were evaluated. GSE39582 was employed to confirmed the levels of genes in CRC compared to the normal samples. Additionally, employing unhealthy samples and the RT-qPCR means our outcomes was validated. Finally, PharmacoGx information were utilized to connect the levels of potential genes to drug tolerance and susceptibility. Our findings showed SMAD2 and SMAD4 levels in TGF-ß signaling were more significant than other pathway genes. Our findings indicated that the protein levels of these genes were lower in malignant tissues than in healthy tissues. Results revealed a significant correlation between low levels of SMAD2 and unfavorable OS in CRC individuals. RT-qPCR results demonstrated decreased expressions of both SMAD2 and SMAD4 in cancer tissues compared to elevated levels in adjacent normal samples. Our results showed significant association between selected genes and immune cell infiltration markers such as CD8+, and B-cells. Our results indicated a potential association among the levels of SMAD2 and SMAD4 genes and tolerance and susceptibility to Nilotinib and Panobinostat drugs. CONCLUSION: Reduced expression of SMAD2 and SMAD4 may be pivotal in CRC progression, impacting downstream genes unrelated to patient OS. These findings suggest a potential role for SMAD2 and SMAD4 as predictive markers for drug response in CRC patients.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs colorectales , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Protéine Smad2 , Protéine Smad-4 , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Protéine Smad-4/génétique , Protéine Smad-4/métabolisme , Protéine Smad2/génétique , Protéine Smad2/métabolisme , Pronostic , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Transduction du signal/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Régulation négative/génétique , Régulation négative/effets des médicaments et des substances chimiques , Femelle
9.
Matrix Biol ; 132: 87-97, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39019241

RÉSUMÉ

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) represents a severe and progressive manifestation of idiopathic interstitial pneumonia marked by an uncertain etiology along with an unfavorable prognosis. Osteoglycin (OGN), belonging to the small leucine-rich proteoglycans family, assumes pivotal functions in both tissue formation and damage response. However, the roles and potential mechanisms of OGN in the context of lung fibrosis remain unexplored. METHODS: The assessment of OGN expression levels in fibrotic lungs was conducted across various experimental lung fibrosis mouse models. To elucidate the effects of OGN on the differentiation of lung myofibroblasts, both OGN knockdown and OGN overexpression were employed in vitro. The expression of integrin αv, along with its colocalization with lysosomes and latency-associated peptide (LAP), was monitored in OGN-knockdown lung myofibroblasts. Furthermore, the role of OGN in lung fibrosis was investigated through OGN knockdown utilizing adeno-related virus serotype 6 (AAV6)-mediated delivery. RESULTS: OGN exhibited upregulation in both lungs and myofibroblasts across diverse lung fibrosis mouse models. And laboratory experiments in vitro demonstrated that OGN knockdown inhibited the TGF-ß/Smad signaling pathway in lung myofibroblasts. Conversely, OGN overexpression promoted TGF-ß/Smad pathway in these cells. Mechanistic insights revealed that OGN knockdown facilitated lysosome-mediated degradation of integrin αv while inhibiting its binding to latency-associated peptide (LAP). Remarkably, AAV6-targeted OGN knockdown ameliorated the extent of lung fibrosis in experimental mouse models. CONCLUSION: Our results indicate that inhibiting OGN signaling could serve as a promising therapeutic way for lung fibrosis.


Sujet(s)
Modèles animaux de maladie humaine , Fibrose pulmonaire idiopathique , Intégrine alphaV , Poumon , Myofibroblastes , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Souris , Intégrine alphaV/métabolisme , Intégrine alphaV/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/génétique , Poumon/métabolisme , Poumon/anatomopathologie , Protéines Smad/métabolisme , Protéines Smad/génétique , Humains , Techniques de knock-down de gènes , Mâle , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/génétique , Fibrose pulmonaire/anatomopathologie
10.
Theriogenology ; 227: 60-67, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39018835

RÉSUMÉ

Follicular wave synchronization and follicular superstimulation with FSH are commonly used in OPU-IVP programs to increase oocyte developmental competence. Factors like Growth Differentiation Factor 9 (GDF9) and Bone Morphogenetic Protein 15 (BMP15), from the TGF beta superfamily, are produced by the oocyte and modulate follicular function. The aim of this study was to analyze the FSH-induced effects on (1) embryo production in dual-purpose Simmental cattle, and (2) TGF beta-mediated effects on oocyte-granulosa cell communication. Simmental heifers (n = 12, age 484 ± 62 days) underwent two OPU-IVP cycles in a cross-over design. Follicular waves were synchronized using 0.5 mg cloprostenol on Day 0, followed by 10 µg buserelin on Day 2. Subsequently, half of the heifers were randomly assigned to receive FSH/LH (four injections of 75 IU FSHp and 75IU LHp, 12 h apart on Days 4 and 5) before the first OPU, while the remaining heifers received FSH/LH before the second OPU. At the time of OPU, i.e. 7 days after the start of synchronization, granulosa cells were collected for RT-qPCR analysis. FSH treatment did not affect the number of oocytes collected (17.3 vs. 13.3, P > 0.05), but increased the percentage of quality 1 oocytes compared to controls (45.7 % vs. 22.0 %, P < 0.001). Neither cleavage (86.4 % vs. 85.7 %), nor blastocyst (42.1 % vs. 39.3 %) rate, or the number of transferable embryos produced by IVP (4.1 vs 4.8) was influenced by FSH treatment (P > 0.05 in all cases). FSH treatment increased HIF1A and FSHR levels in granulosa cells, while STAR was decreased (P = 0.008 in all cases). FSH treatment did not affect BMP15 or GDF9 mRNA expression (P > 0.05) but appeared to modulate the expression of genes involved in the BMP signaling pathway. Transcriptional levels of BMP15 receptor (BMPR1A, P = 0.016), and its downstream signaling factor SMAD1 (P = 0.008) were affected by FSH treatment. Our results demonstrated no benefit of this FSH stimulation protocol on IVP results in Simmental heifers. Further, our results suggest that the effects of FSH on bovine oocytes during acquisition of developmental competence may be mediated through BMP, but do not involve the regulation of transcriptional availability of GDF9, providing new insights into possible paracrine effects of the oocyte on granulosa cells.


Sujet(s)
Hormone folliculostimulante , Cellules de la granulosa , Hormone lutéinisante , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Bovins , Femelle , Cellules de la granulosa/effets des médicaments et des substances chimiques , Cellules de la granulosa/métabolisme , Hormone folliculostimulante/pharmacologie , Hormone folliculostimulante/administration et posologie , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/pharmacologie , Facteur de croissance transformant bêta/métabolisme , Hormone lutéinisante/pharmacologie , Hormone lutéinisante/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Techniques de maturation in vitro des ovocytes/médecine vétérinaire , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Fécondation in vitro/médecine vétérinaire , Synchronisation de l'oestrus
11.
Semin Cancer Biol ; 102-103: 4-16, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38917876

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) is a major contributor to metastatic progression and is prominently regulated by TGF-ß signalling. Both EMT and TGF-ß pathway components are tightly controlled by non-coding RNAs - including microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) - that collectively have major impacts on gene expression and resulting cellular states. While miRNAs are the best characterised regulators of EMT and TGF-ß signaling and the miR-200-ZEB1/2 feedback loop plays a central role, important functions for lncRNAs and circRNAs are also now emerging. This review will summarise our current understanding of the roles of non-coding RNAs in EMT and TGF-ß signaling with a focus on their functions in cancer progression.


Sujet(s)
Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , microARN , Tumeurs , Transduction du signal , Facteur de croissance transformant bêta , Transition épithélio-mésenchymateuse/génétique , Humains , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Animaux , microARN/génétique , ARN long non codant/génétique , ARN circulaire/génétique
12.
Adv Sci (Weinh) ; 11(32): e2401492, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38932472

RÉSUMÉ

Genetic and epigenetic alterations are cancer hallmark characteristics. However, the role of inherited cancer predisposition alleles in co-opting lineage factor epigenetic reprogramming and tumor progression remains elusive. Here the FinnGen cohort phenome-wide analysis, along with multiple genome-wide association studies, has consistently identified the rs339331-RFX6/6q22 locus associated with prostate cancer (PCa) risk across diverse populations. It is uncovered that rs339331 resides in a reprogrammed androgen receptor (AR) binding site in PCa tumors, with the T risk allele enhancing AR chromatin occupancy. RFX6, an AR-regulated gene linked to rs339331, exhibits synergistic prognostic value for PCa recurrence and metastasis. This comprehensive in vitro and in vivo studies demonstrate the oncogenic functions of RFX6 in promoting PCa cell proliferation and metastasis. Mechanistically, RFX6 upregulates HOXA10 that profoundly correlates with adverse PCa outcomes and is pivotal in RFX6-mediated PCa progression, facilitating the epithelial-mesenchymal transition (EMT) and modulating the TGFß/SMAD signaling axis. Clinically, HOXA10 elevation is associated with increased EMT scores, tumor advancement and PCa recurrence. Remarkably, reducing RFX6 expression restores enzalutamide sensitivity in resistant PCa cells and tumors. This findings reveal a complex interplay of genetic and epigenetic mechanisms in PCa pathogenesis and drug resistance, centered around disrupted prostate lineage AR signaling and abnormal RFX6 expression.


Sujet(s)
Allèles , Évolution de la maladie , Résistance aux médicaments antinéoplasiques , Protéines à homéodomaine , Tumeurs de la prostate , Facteurs de transcription des facteurs régulateurs X , Transduction du signal , Facteur de croissance transformant bêta , Mâle , Humains , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Transduction du signal/génétique , Facteurs de transcription des facteurs régulateurs X/génétique , Facteurs de transcription des facteurs régulateurs X/métabolisme , Animaux , Souris , Résistance aux médicaments antinéoplasiques/génétique , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Lignée cellulaire tumorale , Étude d'association pangénomique/méthodes , Modèles animaux de maladie humaine
13.
Reprod Toxicol ; 128: 108650, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38945500

RÉSUMÉ

BACKGROUND: In utero cigarette smoking/nicotine exposure during pregnancy significantly affects fetal development and increases the risk of cardiovascular disease late in life. However, the underlying molecular mechanisms remain largely unknown. We tested the hypothesis that fetal nicotine aerosol exposure reprograms ischemia-sensitive gene expressions, resulting in increased heart susceptibility to ischemic injury and cardiac dysfunction in adulthood. METHODS: Pregnant rats were exposed to chronic intermittent nicotine aerosol (CINA) or saline aerosol control from gestational day 4 to day 21. Experiments were performed on 6-month-old adult offspring. RESULTS: CINA exposure increased ischemia-induced cardiac injury and cardiac dysfunction compared to the control group, which was associated with over- expression of angiotensin II receptor (ATR) protein in the left ventricle (LV) of adult offspring. Meanwhile, CINA exposure up-regulated cardiac TGF-ß/SMADs family proteins in the LV. In addition, CINA exposure enhanced cardiac reactive oxygen species (ROS) production and increased the DNA methylation level. The levels of phosphorylated-Akt were upregulated but LC3B-II/I protein abundances were downregulated in the hearts isolated from the CINA-treated group. CONCLUSION: Fetal nicotine aerosol exposure leads to cardiac dysfunction in response to ischemic stimulation in adulthood. Two molecular pathways are implicated. First, fetal CINA exposure elevates cardiac ATR levels, affecting the TGFß-SMADs pathway. Second, heightened Angiotensin II/ATR signaling triggers ROS production, leading to DNA hypermethylation, p-Akt activation, and autophagy deficiency. These molecular shifts in cardiomyocytes result in the development of a heart ischemia-sensitive phenotype and subsequent dysfunction in adult offspring.


Sujet(s)
Aérosols , Nicotine , Effets différés de l'exposition prénatale à des facteurs de risque , Protéines proto-oncogènes c-akt , Espèces réactives de l'oxygène , Récepteurs aux angiotensines , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Grossesse , Nicotine/toxicité , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Effets différés de l'exposition prénatale à des facteurs de risque/induit chimiquement , Protéines proto-oncogènes c-akt/métabolisme , Espèces réactives de l'oxygène/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Récepteurs aux angiotensines/métabolisme , Récepteurs aux angiotensines/génétique , Rat Sprague-Dawley , Ischémie myocardique/induit chimiquement , Mâle
14.
PLoS Genet ; 20(6): e1011324, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38875298

RÉSUMÉ

The Transforming Growth Factor beta (TGF-ß) family consists of numerous secreted peptide growth factors that play significant roles in cell function, tissue patterning, and organismal homeostasis, including wound repair and immunity. Typically studied as homodimers, these ligands have the potential to diversify their functions through ligand interactions that may enhance, repress, or generate novel functions. In the nematode Caenorhabditis elegans, there are only five TGF-ß ligands, providing an opportunity to dissect ligand interactions in fewer combinations than in vertebrates. As in vertebrates, these ligands can be divided into bone morphogenetic protein (BMP) and TGF-ß/Activin subfamilies that predominantly signal through discrete signaling pathways. The BMP subfamily ligand DBL-1 has been well studied for its role in the innate immune response in C. elegans. Here we show that all five TGF-ß ligands play a role in survival on bacterial pathogens. We also demonstrate that multiple TGF-ß ligand pairs act nonredundantly as part of this response. We show that the two BMP-like ligands-DBL-1 and TIG-2-function independently of each other in the immune response, while TIG-2/BMP and the TGF-ß/Activin-like ligand TIG-3 function together. Structural modeling supports the potential for TIG-2 and TIG-3 to form heterodimers. Additionally, we identify TIG-2 and TIG-3 as members of a rare subset of TGF-ß ligands lacking the conserved cysteine responsible for disulfide linking mature dimers. Finally, we show that canonical DBL-1/BMP receptor and Smad signal transducers function in the response to bacterial pathogens, while components of the DAF-7 TGF-ß/Activin signaling pathway do not play a major role in survival. These results demonstrate a novel potential for BMP and TGF-ß/Activin subfamily ligands to interact and may provide a mechanism for distinguishing the developmental and homeostatic functions of these ligands from an acute response such as the innate immune response to bacterial pathogens.


Sujet(s)
Protéines morphogénétiques osseuses , Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Immunité innée , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Caenorhabditis elegans/microbiologie , Caenorhabditis elegans/génétique , Caenorhabditis elegans/immunologie , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Protéines de Caenorhabditis elegans/génétique , Protéines de Caenorhabditis elegans/métabolisme , Immunité innée/génétique , Ligands , Protéines morphogénétiques osseuses/métabolisme , Protéines morphogénétiques osseuses/génétique , Activines/métabolisme , Activines/génétique , Neuropeptides
15.
Nat Genet ; 56(7): 1468-1481, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38839885

RÉSUMÉ

Aneuploidy is frequently detected in early human embryos as a major cause of early pregnancy failure. However, how aneuploidy affects cellular function remains elusive. Here, we profiled the transcriptomes of 14,908 single cells from 203 human euploid and aneuploid blastocysts involving autosomal and sex chromosomes. Nearly all of the blastocysts contained four lineages. In aneuploid chromosomes, 19.5% ± 1.2% of the expressed genes showed a dosage effect, and 90 dosage-sensitive domains were identified. Aneuploidy leads to prevalent genome-wide transcriptome alterations. Common effects, including apoptosis, were identified, especially in monosomies, partially explaining the lower cell numbers in autosomal monosomies. We further identified lineage-specific effects causing unstable epiblast development in aneuploidies, which was accompanied by the downregulation of TGF-ß and FGF signaling, which resulted in insufficient trophectoderm maturation. Our work provides crucial insights into the molecular basis of human aneuploid blastocysts and may shed light on the cellular interaction during blastocyst development.


Sujet(s)
Aneuploïdie , Blastocyste , Analyse sur cellule unique , Transcriptome , Humains , Blastocyste/métabolisme , Blastocyste/cytologie , Analyse sur cellule unique/méthodes , Femelle , Régulation de l'expression des gènes au cours du développement , Développement embryonnaire/génétique , Analyse de profil d'expression de gènes/méthodes , Grossesse , Transduction du signal/génétique , Apoptose/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Lignage cellulaire/génétique
16.
Mol Biol Rep ; 51(1): 728, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38861185

RÉSUMÉ

INTRODUCTION: Colorectal cancer (CRC) is the second common cancer and the fourth major reason of cancer death worldwide. Dysregulation of intracellular pathways, such as TGF-ß/SMAD signaling, contributes to CRC development. MicroRNAs (miRNAs) are post-transcriptional regulators that are involved in CRC pathogenesis. Here, we aimed to investigate the effect of miR-3613-3p on the TGF-ß /SMAD signaling pathway in CRC. METHODS & RESULTS: Bioinformatics analysis suggested that miR-3613-3p is a regulator of TGF-Β signaling downstream genes. Then, miR-3613-3p overexpression was followed by downregulation of TGF-ßR1, TGF-ßR2, and SMAD2 expression levels, detected by RT-qPCR. Additionally, dual luciferase assay supported the direct interaction of miR-3613-3p with 3'UTR sequences of TGF-ßR1 and TGF-ßR2 genes. Furthermore, reduced SMAD3 protein level following the miR-3613-3p overexpression verified its suppressive effect against TGF-ß signaling in HCT-116 cells, detected by western blot analysis. Finally, miR-3613-3p overexpression induced sub-G1 arrest in HCT116 cells, detected by flow cytometry, and promoted downregulation of cyclin D1 protein expression, which was detected by western blotting analysis. CONCLUSION: Our findings indicated that miR-3613-3p plays an important role in CRC by targeting the TGF-ß/SMAD signaling pathway and could be considered as a new candidate for further therapy investigations.


Sujet(s)
Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , microARN , Transduction du signal , Facteur de croissance transformant bêta , Humains , microARN/génétique , microARN/métabolisme , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Transduction du signal/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Régulation de l'expression des gènes tumoraux/génétique , Cellules HCT116 , Récepteur de type I du facteur de croissance transformant bêta/génétique , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Récepteur de type II du facteur de croissance transformant bêta/génétique , Récepteur de type II du facteur de croissance transformant bêta/métabolisme , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Prolifération cellulaire/génétique , Régions 3' non traduites/génétique , Lignée cellulaire tumorale , Protéine Smad-3/génétique , Protéine Smad-3/métabolisme
17.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(3): 384-389, 2024 Jun 18.
Article de Chinois | MEDLINE | ID: mdl-38864121

RÉSUMÉ

OBJECTIVE: To explore the association between polymorphisms of transforming growth factor-ß (TGF-ß) signaling pathway and non-syndromic cleft lip with or without cleft palate (NSCL/P) among Asian populations, while considering gene-gene interaction and gene-environment interaction. METHODS: A total of 1 038 Asian NSCL/P case-parent trios were ascertained from an international consortium, which conducted a genome-wide association study using a case-parent trio design to investigate the genes affec-ting risk to NSCL/P. After stringent quality control measures, 343 single nucleotide polymorphism (SNP) spanning across 10 pivotal genes in the TGF-ß signaling pathway were selected from the original genome-wide association study(GWAS) dataset for further analysis. The transmission disequilibrium test (TDT) was used to test for SNP effects. The conditional Logistic regression models were used to test for gene-gene interaction and gene-environment interaction. Environmental factors collected for the study included smoking during pregnancy, passive smoking during pregnancy, alcohol intake during pregnancy, and vitamin use during pregnancy. Due to the low rates of exposure to smoking during pregnancy and alcohol consumption during pregnancy (<3%), only the interaction between maternal smoking during pregnancy and multivitamin supplementation during pregnancy was analyzed. The threshold for statistical significance was rigorously set at P =1.46×10-4, applying Bonferroni correction to account for multiple testing. RESULTS: A total of 23 SNPs in 4 genes yielded nominal association with NSCL/P (P<0.05), but none of these associations was statistically significant after Bonferroni' s multiple test correction. However, there were 6 pairs of SNPs rs4939874 (SMAD2) and rs1864615 (TGFBR2), rs2796813 (TGFB2) and rs2132298 (TGFBR2), rs4147358 (SMAD3) and rs1346907 (TGFBR2), rs4939874 (SMAD2) and rs1019855 (TGFBR2), rs4939874 (SMAD2) and rs12490466 (TGFBR2), rs2009112 (TGFB2) and rs4075748 (TGFBR2) showed statistically significant SNP-SNP interaction (P<1.46×10-4). In contrast, the analysis of gene-environment interactions did not yield any significant results after being corrected by multiple testing. CONCLUSION: The comprehensive evaluation of SNP associations and interactions within the TGF-ß signaling pathway did not yield any direct associations with NSCL/P risk in Asian populations. However, the significant gene-gene interactions identified suggest that the genetic architecture influencing NSCL/P risk may involve interactions between genes within the TGF-ß signaling pathway. These findings underscore the necessity for further investigations to unravel these results and further explore the underlying biological mechanisms.


Sujet(s)
Bec-de-lièvre , Fente palatine , Interaction entre gènes et environnement , Étude d'association pangénomique , Polymorphisme de nucléotide simple , Transduction du signal , Facteur de croissance transformant bêta , Humains , Fente palatine/génétique , Bec-de-lièvre/génétique , Transduction du signal/génétique , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/métabolisme , Femelle , Asiatiques/génétique , Grossesse , Mâle , Prédisposition génétique à une maladie , Protéine Smad-3/génétique , Facteurs de risque , Protéine Smad2/génétique , Protéine Smad2/métabolisme , Épistasie , Pollution par la fumée de tabac/effets indésirables , Consommation d'alcool/génétique
18.
Adv Exp Med Biol ; 1441: 885-900, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884757

RÉSUMÉ

The process of valve formation is a complex process that involves intricate interplay between various pathways at precise times. Although we have not completely elucidated the molecular pathways that lead to normal valve formation, we have identified a few major players in this process. We are now able to implicate TGF-ß, BMP, and NOTCH as suspects in tricuspid atresia (TA), as well as their downstream targets: NKX2-5, TBX5, NFATC1, GATA4, and SOX9. We know that the TGF-ß and the BMP pathways converge on the SMAD4 molecule, and we believe that this molecule plays a very important role to tie both pathways to TA. Similarly, we look at the NOTCH pathway and identify the HEY2 as a potential link between this pathway and TA. Another transcription factor that has been implicated in TA is NFATC1. While several mouse models exist that include part of the TA abnormality as their phenotype, no true mouse model can be said to represent TA. Bridging this gap will surely shed light on this complex molecular pathway and allow for better understanding of the disease process.


Sujet(s)
Modèles animaux de maladie humaine , Transduction du signal , Atrésie tricuspide , Animaux , Atrésie tricuspide/génétique , Atrésie tricuspide/métabolisme , Atrésie tricuspide/anatomopathologie , Humains , Souris , Cœur univentriculaire/génétique , Cœur univentriculaire/métabolisme , Cœur univentriculaire/physiopathologie , Cœur univentriculaire/anatomopathologie , Facteurs de transcription NFATC/métabolisme , Facteurs de transcription NFATC/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique
19.
Reprod Fertil Dev ; 362024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38894494

RÉSUMÉ

Context Altered signalling of androgens, anti-Müllerian hormone or transforming growth factor beta (TGFß) during foetal development have been implicated in the predisposition to polycystic ovary syndrome (PCOS) in later life, aside from its genetic predisposition. In foetal ovarian fibroblasts, TGFß1 has been shown to regulate androgen signalling and seven genes located in loci associated with PCOS. Since PCOS exhibits a myriad of symptoms, it likely involves many different organs. Aims To identify the relationships between TGFß signalling molecules and PCOS candidate genes in different tissues associated with PCOS. Methods Using RNA sequencing data, we examined the expression patterns of TGFß signalling molecules in the human ovary, testis, heart, liver, kidney, brain tissue, and cerebellum from 4 to 20weeks of gestation and postnatally. We also examined the correlations between gene expression of TGFß signalling molecules and PCOS candidate genes. Key results TGFß signalling molecules were dynamically expressed in most tissues prenatally and/or postnatally. FBN3 , a PCOS candidate gene involved in TGFß signalling, was expressed during foetal development in all tissues. The PCOS candidate genes HMGA2, YAP1 , and RAD50 correlated significantly (P TGFBR1 in six out of the seven tissues examined. Conclusions This study suggests that possible crosstalk occurs between genes in loci associated with PCOS and TGFß signalling molecules in multiple tissues, particularly during foetal development. Implications Thus, alteration in TGFß signalling during foetal development could affect many tissues contributing to the multiple phenotypes of PCOS in later life.


Sujet(s)
Syndrome des ovaires polykystiques , Transduction du signal , Facteur de croissance transformant bêta , Humains , Femelle , Syndrome des ovaires polykystiques/génétique , Syndrome des ovaires polykystiques/métabolisme , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Adulte , Ovaire/métabolisme , Foetus/métabolisme , Mâle , Grossesse , Régulation de l'expression des gènes au cours du développement , Testicule/métabolisme , Testicule/embryologie , Fibrillines
20.
Folia Med (Plovdiv) ; 66(2): 243-249, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38690820

RÉSUMÉ

AIM: The aim of this study was to evaluate specific single nucleotide polymorphisms (SNP) of transforming growth factor-beta (TGF-ß) (rs1800469) and insulin-like growth factor-1 (IGF-1) (rs17032362) genes in Class II individuals with a normal maxilla and retrognathic (short) mandible.


Sujet(s)
Facteur de croissance IGF-I , Malocclusion de classe II , Mandibule , Facteur de croissance transformant bêta , Adolescent , Adulte , Femelle , Humains , Mâle , Facteur de croissance IGF-I/génétique , Malocclusion de classe II/génétique , Polymorphisme de nucléotide simple , Rétrognathie/génétique , Facteur de croissance transformant bêta/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE