Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.042
Filtrer
1.
Development ; 151(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38828908

RÉSUMÉ

During limb bud formation, axis polarities are established as evidenced by the spatially restricted expression of key regulator genes. In particular, the mutually antagonistic interaction between the GLI3 repressor and HAND2 results in distinct and non-overlapping anterior-distal Gli3 and posterior Hand2 expression domains. This is a hallmark of the establishment of antero-posterior limb axis polarity, together with spatially restricted expression of homeodomain and other transcriptional regulators. Here, we show that TBX3 is required for establishment of the posterior expression boundary of anterior genes in mouse limb buds. ChIP-seq and differential gene expression analysis of wild-type and mutant limb buds identifies TBX3-specific and shared TBX3-HAND2 target genes. High sensitivity fluorescent whole-mount in situ hybridisation shows that the posterior expression boundaries of anterior genes are positioned by TBX3-mediated repression, which excludes anterior genes such as Gli3, Alx4, Hand1 and Irx3/5 from the posterior limb bud mesenchyme. This exclusion delineates the posterior mesenchymal territory competent to establish the Shh-expressing limb bud organiser. In turn, HAND2 is required for Shh activation and cooperates with TBX3 to upregulate shared posterior identity target genes in early limb buds.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice , Régulation de l'expression des gènes au cours du développement , Bourgeons de membre , Protéines à domaine boîte-T , Animaux , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Bourgeons de membre/métabolisme , Bourgeons de membre/embryologie , Souris , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Protéine à doigts de zinc Gli3/métabolisme , Protéine à doigts de zinc Gli3/génétique , Régulation positive/génétique , Plan d'organisation du corps/génétique , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Mésoderme/métabolisme , Mésoderme/embryologie
2.
Curr Top Dev Biol ; 159: 232-271, 2024.
Article de Anglais | MEDLINE | ID: mdl-38729677

RÉSUMÉ

The anterior-to-posterior (head-to-tail) body axis is extraordinarily diverse among vertebrates but conserved within species. Body axis development requires a population of axial progenitors that resides at the posterior of the embryo to sustain elongation and is then eliminated once axis extension is complete. These progenitors occupy distinct domains in the posterior (tail-end) of the embryo and contribute to various lineages along the body axis. The subset of axial progenitors with neuromesodermal competency will generate both the neural tube (the precursor of the spinal cord), and the trunk and tail somites (producing the musculoskeleton) during embryo development. These axial progenitors are called Neuromesodermal Competent cells (NMCs) and Neuromesodermal Progenitors (NMPs). NMCs/NMPs have recently attracted interest beyond the field of developmental biology due to their clinical potential. In the mouse, the maintenance of neuromesodermal competency relies on a fine balance between a trio of known signals: Wnt/ß-catenin, FGF signalling activity and suppression of retinoic acid signalling. These signals regulate the relative expression levels of the mesodermal transcription factor Brachyury and the neural transcription factor Sox2, permitting the maintenance of progenitor identity when co-expressed, and either mesoderm or neural lineage commitment when the balance is tilted towards either Brachyury or Sox2, respectively. Despite important advances in understanding key genes and cellular behaviours involved in these fate decisions, how the balance between mesodermal and neural fates is achieved remains largely unknown. In this chapter, we provide an overview of signalling and gene regulatory networks in NMCs/NMPs. We discuss mutant phenotypes associated with axial defects, hinting at the potential significant role of lesser studied proteins in the maintenance and differentiation of the progenitors that fuel axial elongation.


Sujet(s)
Plan d'organisation du corps , Mésoderme , Animaux , Plan d'organisation du corps/génétique , Mésoderme/métabolisme , Mésoderme/cytologie , Mésoderme/embryologie , Régulation de l'expression des gènes au cours du développement , Humains , Transduction du signal , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Différenciation cellulaire , Tête/embryologie
3.
Nat Commun ; 15(1): 4108, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750011

RÉSUMÉ

MAPK pathway-driven tumorigenesis, often induced by BRAFV600E, relies on epithelial dedifferentiation. However, how lineage differentiation events are reprogrammed remains unexplored. Here, we demonstrate that proteostatic reactivation of developmental factor, TBX3, accounts for BRAF/MAPK-mediated dedifferentiation and tumorigenesis. During embryonic development, BRAF/MAPK upregulates USP15 to stabilize TBX3, which orchestrates organogenesis by restraining differentiation. The USP15-TBX3 axis is reactivated during tumorigenesis, and Usp15 knockout prohibits BRAFV600E-driven tumor development in a Tbx3-dependent manner. Deleting Tbx3 or Usp15 leads to tumor redifferentiation, which parallels their overdifferentiation tendency during development, exemplified by disrupted thyroid folliculogenesis and elevated differentiation factors such as Tpo, Nis, Tg. The clinical relevance is highlighted in that both USP15 and TBX3 highly correlates with BRAFV600E signature and poor tumor prognosis. Thus, USP15 stabilized TBX3 represents a critical proteostatic mechanism downstream of BRAF/MAPK-directed developmental homeostasis and pathological transformation, supporting that tumorigenesis largely relies on epithelial dedifferentiation achieved via embryonic regulatory program reinitiation.


Sujet(s)
Carcinogenèse , Protéines proto-oncogènes B-raf , Protéines à domaine boîte-T , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Animaux , Humains , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Souris , Différenciation cellulaire , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Système de signalisation des MAP kinases/génétique , Régulation de l'expression des gènes tumoraux , Souris knockout , Femelle , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme
4.
Biochem Biophys Res Commun ; 718: 150037, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38735135

RÉSUMÉ

BACKGROUND: Non-small cell lung cancer (NSCLC) accounts for more than 80 % of lung cancer (LC) cases, making it the primary cause of cancer-related mortality worldwide. T-box transcription factor 5 (TBX5) is an important regulator of embryonic and organ development and plays a key role in cancer development. Here, our objective was to investigate the involvement of TBX5 in ferroptosis within LC cells and the underlying mechanisms. METHODS: First, TBX5 expression was examined in human LC cells. Next, overexpression of TBX5 and Yes1-associated transcriptional regulator (YAP1) and knockdown of TEA domain 1 (TEAD1) were performed in A549 and NCI-H1703 cells. The proliferation ability of A549 and NCI-H1703 cells, GSH, MDA, ROS, and Fe2+ levels were measured. Co-immunoprecipitation (Co-IP) was performed to verify whether TBX5 protein could bind YAP1. Then TBX5, YAP1, TEAD1, GPX4, p53, FTH1, SLC7A11 and PTGS2 protein levels were assessed. Finally, we verified the effect of TBX5 on ferroptosis in LC cells in vivo. RESULTS: TBX5 expression was down-regulated in LC cells, especially in A549 and NCI-H1703 cells. Overexpression of TBX5 significantly decreased proliferation ability of A549 and NCI-H1703 cells, downregulated GPX4 and GSH levels, and upregulated MDA, ROS, and Fe2+ levels. Co-IP verified that TBX5 protein could bind YAP1. Moreover, oe-YAP1 promoted proliferation ability of A549 and NCI-H1703 cells transfected with Lv-TBX5, upregulated GPX4 and GSH levels and downregulated MDA, ROS, and Fe2+ levels. Additionally, oe-YAP1 promoted FTH1 and SLC7A11 levels and inhibited p53 and PTGS2 levels in A549 and NCI-H1703 cells transfected with Lv-TBX5. However, transfection with si-TEAD1 further reversed these effects. In vivo experiments further validated that TBX5 promoted ferroptosis in LC cells. CONCLUSIONS: TBX5 inhibited the activation of YAP1-TEAD1 pathway to promote ferroptosis in LC cells.


Sujet(s)
Ferroptose , Tumeurs du poumon , Protéines à domaine boîte-T , Facteurs de transcription à domaine TEA , Facteurs de transcription , Protéines de signalisation YAP , Ferroptose/génétique , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Protéines de signalisation YAP/métabolisme , Protéines de signalisation YAP/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription à domaine TEA/métabolisme , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Animaux , Lignée cellulaire tumorale , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Souris nude , Prolifération cellulaire , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Souris , Régulation de l'expression des gènes tumoraux , Cellules A549 , Transduction du signal , Espèces réactives de l'oxygène/métabolisme
5.
Am J Physiol Cell Physiol ; 326(5): C1384-C1397, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38690917

RÉSUMÉ

Metabolic dysfunction of the extracellular matrix (ECM) is one of the primary causes of intervertebral disc degeneration (IVDD). Previous studies have demonstrated that the transcription factor Brachyury (Bry) has the potential to promote the synthesis of collagen II and aggrecan, while the specific mechanism is still unknown. In this study, we used a lipopolysaccharide (LPS)-induced model of nucleus pulposus cell (NPC) degeneration and a rat acupuncture IVDD model to elucidate the precise mechanism through which Bry affects collagen II and aggrecan synthesis in vitro and in vivo. First, we confirmed Bry expression decreased in degenerated human nucleus pulposus (NP) cells (NPCs). Knockdown of Bry exacerbated the decrease in collagen II and aggrecan expression in the lipopolysaccharide (LPS)-induced NPCs degeneration in vitro model. Bioinformatic analysis indicated that Smad3 may participate in the regulatory pathway of ECM synthesis regulated by Bry. Chromatin immunoprecipitation followed by quantitative polymerase chain reaction (ChIP-qPCR) and luciferase reporter gene assays demonstrated that Bry enhances the transcription of Smad3 by interacting with a specific motif on the promoter region. In addition, Western blot and reverse transcription-qPCR assays demonstrated that Smad3 positively regulates the expression of aggrecan and collagen II in NPCs. The following rescue experiments revealed that Bry-mediated regulation of ECM synthesis is partially dependent on Smad3 phosphorylation. Finally, the findings from the in vivo rat acupuncture-induced IVDD model were consistent with those obtained from in vitro assays. In conclusion, this study reveals that Bry positively regulates the synthesis of collagen II and aggrecan in NP through transcriptional activation of Smad3.NEW & NOTEWORTHY Mechanically, in the nucleus, Bry enhances the transcription of Smad3, leading to increased expression of Smad3 protein levels; in the cytoplasm, elevated substrate levels further lead to an increase in the phosphorylation of Smad3, thereby regulating collagen II and aggrecan expression. Further in vivo experiments provide additional evidence that Bry can alleviate IVDD through this mechanism.


Sujet(s)
Agrécanes , Matrice extracellulaire , Protéines foetales , Dégénérescence de disque intervertébral , Nucleus pulposus , Rat Sprague-Dawley , Protéine Smad-3 , Protéines à domaine boîte-T , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Nucleus pulposus/métabolisme , Nucleus pulposus/anatomopathologie , Animaux , Matrice extracellulaire/métabolisme , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Humains , Rats , Dégénérescence de disque intervertébral/génétique , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , Agrécanes/métabolisme , Agrécanes/génétique , Mâle , Protéines foetales/génétique , Protéines foetales/métabolisme , Collagène de type II/métabolisme , Collagène de type II/génétique , Régulation de l'expression des gènes , Femelle , Adulte , Adulte d'âge moyen , Cellules cultivées , Transcription génétique
6.
Neurosurg Focus ; 56(5): E18, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38691860

RÉSUMÉ

Chordomas are tumors thought to originate from notochordal remnants that occur in midline structures from the cloves of the skull base to the sacrum. In adults, the most common location is the sacrum, followed by the clivus and then mobile spine, while in children a clival origin is most common. Most chordomas are slow growing. Clinical presentation of chordomas tend to occur late, with local invasion and large size often complicating surgical intervention. Radiation therapy with protons has been proven to be an effective adjuvant therapy. Unfortunately, few adjuvant systemic treatments have demonstrated significant effectiveness, and chordomas tend to recur despite intensive multimodal care. However, insight into the molecular underpinnings of chordomas may guide novel therapeutic approaches including selection for immune and molecular therapies, individualized prognostication of outcomes, and real-time noninvasive assessment of disease burden and evolution. At the genomic level, elevated levels of brachyury stemming from duplications and mutations resulting in altered transcriptional regulation may introduce druggable targets for new surgical adjuncts. Transcriptome and epigenome profiling have revealed promoter- and enhancer-dependent mechanisms of protein regulation, which may influence therapeutic response and long-term disease history. Continued scientific and clinical advancements may offer further opportunities for treatment of chordomas. Single-cell transcriptome profiling has further provided insight into the heterogeneous molecular pathways contributing to chordoma propagation. New technologies such as spatial transcriptomics and emerging biochemical analytes such as cell-free DNA have further augmented the surgeon-clinician's armamentarium by facilitating detailed characterization of intra- and intertumoral biology while also demonstrating promise for point-of-care tumor quantitation and assessment. Recent and ongoing clinical trials highlight accelerating interest to translate laboratory breakthroughs in chordoma biology and immunology into clinical care. In this review, the authors dissect the landmark studies exploring the molecular pathogenesis of chordoma. Incorporating this into an outline of ongoing clinical trials and discussion of emerging technologies, the authors aimed to summarize recent advancements in understanding chordoma pathogenesis and how neurosurgical care of chordomas may be augmented by improvements in adjunctive treatments.


Sujet(s)
Chordome , Protéines foetales , Chordome/génétique , Chordome/thérapie , Humains , Carcinogenèse/génétique , Protéines à domaine boîte-T/génétique , Tumeurs de la base du crâne/génétique , Tumeurs de la base du crâne/thérapie
7.
Biochem Biophys Res Commun ; 720: 150104, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-38749189

RÉSUMÉ

The T-BOX transcription factor TBX1 is essential for the development of the pharyngeal apparatus and it is haploinsufficient in DiGeorge syndrome (DGS), a developmental anomaly associated with congenital heart disease and other abnormalities. The murine model recapitulates the heart phenotype and showed collagen accumulation. We first used a cellular model to study gene expression during cardiogenic differentiation of WT and Tbx1-/- mouse embryonic stem cells. Then we used a mouse model of DGS to test whether interfering with collagen accumulation using an inhibitor of lysyl hydroxylase would modify the cardiac phenotype of the mutant. We found that loss of Tbx1 in a precardiac differentiation model was associated with up regulation of a subset of ECM-related genes, including several collagen genes. In the in vivo model, early prenatal treatment with Minoxidil, a lysyl hydroxylase inhibitor, ameliorated the cardiac outflow tract septation phenotype in Tbx1 mutant fetuses, but it had no effect on septation in WT fetuses. We conclude that TBX1 suppresses a defined subset of ECM-related genes. This function is critical for OFT septation because the inhibition of collagen cross-linking in the mutant reduces significantly the penetrance of septation defects.


Sujet(s)
Syndrome de DiGeorge , Modèles animaux de maladie humaine , Minoxidil , Protéines à domaine boîte-T , Animaux , Syndrome de DiGeorge/génétique , Syndrome de DiGeorge/métabolisme , Syndrome de DiGeorge/traitement médicamenteux , Syndrome de DiGeorge/anatomopathologie , Souris , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Minoxidil/pharmacologie , Collagène/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques
8.
Cell Rep ; 43(5): 114222, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38735046

RÉSUMÉ

The activation and specialization of regulatory T cells (Tregs) are crucial for maintaining immune self-tolerance; however, the regulation of these processes by histone modifications is not fully understood. Here, we show that T cell-specific deletion of the lysine methyltransferase MLL1 results in a spontaneous lymphocyte proliferation phenotype in aged mice without disturbing the development of conventional T cells and Tregs. Treg-specific MLL1 ablation leads to a systemic autoimmune disease associated with Treg dysfunction. Moreover, RNA sequencing demonstrates that the induction of multiple genes involved in Treg activation, functional specialization, and tissue immigration is defective in MLL1-deficient Tregs. This dysregulation is associated with defects in H3K4 trimethylation at these genes' transcription start sites. Finally, using a T-bet fate-mapping mouse system, we determine that MLL1 is required to establish stable Th1-type Tregs. Thus, MLL1 is essential in optimal Treg function by providing a coordinated chromatin context for activation and specialization.


Sujet(s)
Histone-lysine N-methyltransferase , Activation des lymphocytes , Protéine de la leucémie myéloïde-lymphoïde , Lymphocytes T régulateurs , Animaux , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/métabolisme , Protéine de la leucémie myéloïde-lymphoïde/génétique , Souris , Souris de lignée C57BL , Histone/métabolisme , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Méthylation , Prolifération cellulaire
9.
Stem Cell Res Ther ; 15(1): 148, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38778426

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) are known as one of the best candidate cells to produce cardiac pacemaker-like cells (CPLCs). Upregulation of TBX3 transcription factor and inhibition of the nodal signal pathway have a significant role in the formation of cardiac pacemaker cells such as sinoatrial and atrioventricular nodes, which initiate the heartbeat and control the rhythm of heart contractions. This study aimed to confirm the effects of transfection of TBX3 transcription factor and inhibition of the nodal signal pathway on differentiating adipose-derived MSCs (AD-MSCs) to CPLCs. AD-MSCs were characterized using flow cytometry and three-lineage differentiation staining. METHODS: The transfection of TBX3 plasmid was carried out using lipofectamine, and inhibition of the nodal signal pathway was done using the small-molecule SB431542. The morphology of the cells was observed using a light microscope. Pacemaker-specific markers, including TBX3, Cx30, HCN4, HCN1, HCN3, and KCNN4, were evaluated using the qRT-PCR method. For protein level, TBX3 and Cx30 were evaluated using ELISA and immunofluorescence staining. The electrophysiology of cells was evaluated using a patch clamp. RESULTS: The TBX3 expression in the TBX3, SM, and TBX + SM groups significantly higher (p < 0.05) compared to the control group and cardiomyocytes. The expression of Cx40 and Cx43 genes were lower in TBX3, SM, TBX + SM groups. In contrast, Cx30 gene showed higher expression in TBX3 group. The expression HCN1, HCN3, and HCN4 genes are higher in TBX3 group. CONCLUSION: The transfection of TBX3 and inhibition of the nodal signal pathway by small-molecule SB431542 enhanced differentiation of AD-MSCs to CPLCs.


Sujet(s)
Différenciation cellulaire , Cellules souches mésenchymateuses , Transduction du signal , Protéines à domaine boîte-T , Transfection , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Humains , Myocytes cardiaques/métabolisme , Myocytes cardiaques/cytologie , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Cellules cultivées , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation/génétique , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation/métabolisme
10.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 150-154, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38814224

RÉSUMÉ

We aimed to observe the effects of adipose-derived mesenchymal stem cells (ADSCs) on T helper 17 (Th17)/regulatory T cells (Treg) and T-box transcription factor (T-bet)/GATA-binding protein 3 (GATA-3) in model mice with primary immune thrombocytopenia (ITP). 32 BALB/C mice were selected. ADSCs were isolated from 2 mice and cultured. The other 30 mice were randomly divided into the normal control group, the ITP model control group, and the ITP experimental group. Platelet count (PLT), Th17/Treg cells, related serum cytokines [interleukin-6 (IL-6), IL-17A, IL-10, and transforming growth factor ß1 (TGF-ß1)], T-bet and GATA-3 mRNA levels in peripheral blood mononuclear cells (PBMCs) in the 3 groups were detected. PLT and Treg in the ITP experimental group were significantly lower than those in the normal control group (P<0.05), but significantly higher than those in the ITP model control group (P<0.05). Th17 and Th17/Treg in the ITP experimental group were significantly higher than those in the normal control group (P<0.05), but significantly lower than those in the ITP model control group (P<0.05). Serum IL-6 and IL-17A levels, and T-bet mRNA levels in the ITP experimental group were significantly higher than those in the normal control group (P<0.05), but significantly lower than those in the ITP model control group (P<0.05). Serum IL-10 and TGF-ß levels, and GATA-3 mRNA levels in the ITP experimental group were significantly lower than those in the normal control group (P<0.05), but significantly higher than those in the ITP model control group (P<0.05). ADSCs can effectively regulate Th17/Treg balance and improve T-bet/GATA-3 mRNA expression levels in ITP model mice.


Sujet(s)
Modèles animaux de maladie humaine , Facteur de transcription GATA-3 , Cellules souches mésenchymateuses , Souris de lignée BALB C , Protéines à domaine boîte-T , Lymphocytes T régulateurs , Cellules Th17 , Animaux , Lymphocytes T régulateurs/métabolisme , Lymphocytes T régulateurs/immunologie , Cellules Th17/métabolisme , Cellules Th17/immunologie , Facteur de transcription GATA-3/génétique , Facteur de transcription GATA-3/métabolisme , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Cellules souches mésenchymateuses/métabolisme , Souris , ARN messager/génétique , ARN messager/métabolisme , Purpura thrombopénique idiopathique/sang , Purpura thrombopénique idiopathique/immunologie , Interleukine-6/sang , Interleukine-6/métabolisme , Interleukine-6/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Facteur de croissance transformant bêta-1/sang , Interleukine-10/génétique , Interleukine-10/sang , Interleukine-10/métabolisme , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Numération des plaquettes , Femelle , Interleukine-17/sang , Interleukine-17/métabolisme , Interleukine-17/génétique , Cytokines/métabolisme , Cytokines/sang , Mâle
11.
Nat Commun ; 15(1): 4650, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38821936

RÉSUMÉ

Synovial tissue inflammation is a hallmark of rheumatoid arthritis (RA). Recent work has identified prominent pathogenic cell states in inflamed RA synovial tissue, such as T peripheral helper cells; however, the epigenetic regulation of these states has yet to be defined. Here, we examine genome-wide open chromatin at single-cell resolution in 30 synovial tissue samples, including 12 samples with transcriptional data in multimodal experiments. We identify 24 chromatin classes and predict their associated transcription factors, including a CD8 + GZMK+ class associated with EOMES and a lining fibroblast class associated with AP-1. By integrating with an RA tissue transcriptional atlas, we propose that these chromatin classes represent 'superstates' corresponding to multiple transcriptional cell states. Finally, we demonstrate the utility of this RA tissue chromatin atlas through the associations between disease phenotypes and chromatin class abundance, as well as the nomination of classes mediating the effects of putatively causal RA genetic variants.


Sujet(s)
Polyarthrite rhumatoïde , Chromatine , Membrane synoviale , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/immunologie , Humains , Chromatine/métabolisme , Chromatine/génétique , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Épigenèse génétique , Analyse sur cellule unique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Fibroblastes/métabolisme , Facteur de transcription AP-1/métabolisme , Facteur de transcription AP-1/génétique , Transcription génétique , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme
12.
Am J Hum Genet ; 111(5): 939-953, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38608674

RÉSUMÉ

Changes in gene regulatory elements play critical roles in human phenotypic divergence. However, identifying the base-pair changes responsible for the distinctive morphology of Homo sapiens remains challenging. Here, we report a noncoding single-nucleotide polymorphism (SNP), rs41298798, as a potential causal variant contributing to the morphology of the skull base and vertebral structures found in Homo sapiens. Screening for differentially regulated genes between Homo sapiens and extinct relatives revealed 13 candidate genes associated with basicranial development, with TBX1, implicated in DiGeorge syndrome, playing a pivotal role. Epigenetic markers and in silico analyses prioritized rs41298798 within a TBX1 intron for functional validation. CRISPR editing revealed that the 41-base-pair region surrounding rs41298798 modulates gene expression at 22q11.21. The derived allele of rs41298798 acts as an allele-specific enhancer mediated by E2F1, resulting in increased TBX1 expression levels compared to the ancestral allele. Tbx1-knockout mice exhibited skull base and vertebral abnormalities similar to those seen in DiGeorge syndrome. Phenotypic differences associated with TBX1 deficiency are observed between Homo sapiens and Neanderthals (Homo neanderthalensis). In conclusion, the regulatory divergence of TBX1 contributes to the formation of skull base and vertebral structures found in Homo sapiens.


Sujet(s)
Polymorphisme de nucléotide simple , Protéines à domaine boîte-T , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Humains , Animaux , Souris , Syndrome de DiGeorge/génétique , Néandertaliens/génétique , Souris knockout , Crâne/anatomie et histologie , Allèles , Rachis/anatomie et histologie , Rachis/malformations , Chromosomes humains de la paire 22/génétique , Phénotype
13.
Immunohorizons ; 8(4): 355-362, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38687282

RÉSUMÉ

To defend against intracellular pathogens such as Toxoplasma gondii, the host generates a robust type 1 immune response. Specifically, host defense against T. gondii is defined by an IL-12-dependent IFN-γ response that is critical for host resistance. Previously, we demonstrated that host resistance is mediated by T-bet-dependent ILC-derived IFN-γ by maintaining IRF8+ conventional type 1 dendritic cells during parasitic infection. Therefore, we hypothesized that innate lymphoid cells are indispensable for host survival. Surprisingly, we observed that T-bet-deficient mice succumb to infection quicker than do mice lacking lymphocytes, suggesting an unknown T-bet-dependent-mediated host defense pathway. Analysis of parasite-mediated inflammatory myeloid cells revealed a novel subpopulation of T-bet+ myeloid cells (TMCs). Our results reveal that TMCs have the largest intracellular parasite burden compared with other professional phagocytes, suggesting they are associated with active killing of T. gondii. Mechanistically, we established that IL-12 is necessary for the induction of inflammatory TMCs during infection and these cells are linked to a role in host survival.


Sujet(s)
Interleukine-12 , Souris de lignée C57BL , Souris knockout , Cellules myéloïdes , Protéines à domaine boîte-T , Toxoplasma , Toxoplasmose , Animaux , Toxoplasma/immunologie , Souris , Interleukine-12/métabolisme , Interleukine-12/immunologie , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/immunologie , Cellules myéloïdes/immunologie , Cellules myéloïdes/métabolisme , Toxoplasmose/immunologie , Toxoplasmose/parasitologie , Interféron gamma/métabolisme , Interféron gamma/immunologie , Immunité innée , Toxoplasmose animale/immunologie , Résistance à la maladie/immunologie , Femelle
14.
Dev Cell ; 59(10): 1252-1268.e13, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38579720

RÉSUMÉ

The blueprint of the mammalian body plan is laid out during gastrulation, when a trilaminar embryo is formed. This process entails a burst of proliferation, the ingression of embryonic epiblast cells at the primitive streak, and their priming toward primitive streak fates. How these different events are coordinated remains unknown. Here, we developed and characterized a 3D culture of self-renewing mouse embryonic cells that captures the main transcriptional and architectural features of the early gastrulating mouse epiblast. Using this system in combination with microfabrication and in vivo experiments, we found that proliferation-induced crowding triggers delamination of cells that express high levels of the apical polarity protein aPKC. Upon delamination, cells become more sensitive to Wnt signaling and upregulate the expression of primitive streak markers such as Brachyury. This mechanistic coupling between ingression and differentiation ensures that the right cell types become specified at the right place during embryonic development.


Sujet(s)
Différenciation cellulaire , Gastrulation , Feuillets embryonnaires , Animaux , Souris , Feuillets embryonnaires/cytologie , Feuillets embryonnaires/métabolisme , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , Ligne primitive/cytologie , Ligne primitive/métabolisme , Protéines foetales/métabolisme , Protéines foetales/génétique , Voie de signalisation Wnt , Prolifération cellulaire , Régulation de l'expression des gènes au cours du développement , Embryon de mammifère/cytologie , Embryon de mammifère/métabolisme
15.
Int J Biol Macromol ; 268(Pt 2): 130853, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38570000

RÉSUMÉ

BACKGROUND: Pulmonary hypertension (PH) is a complex vascular disorder, characterized by pulmonary vessel remodeling and perivascular inflammation. Pulmonary arterial smooth muscle cells (PASMCs) pyroptosis is a novel pathological mechanism implicated of pulmonary vessel remodeling. However, the involvement of circRNAs in the process of pyroptosis and the underlying regulatory mechanisms remain inadequately understood. METHODS: Western blotting, PI staining and LDH release were used to explore the role of circLrch3 in PASMCs pyroptosis. Moreover, S9.6 dot blot and DRIP-PCR were used to assess the formation of R-loop between circLrch3 and its host gene Lrch3. Chip-qPCR were used to evaluate the mechanism of super enhancer-associated circLrh3, which is transcriptionally activated by the transcription factor Tbx2. RESULTS: CircLrch3 was markedly upregulated in hypoxic PASMCs. CircLrch3 knockdown inhibited hypoxia induced PASMCs pyroptosis in vivo and in vitro. Mechanistically, circLrch3 can form R-loop with host gene to upregulate the protein and mRNA expression of Lrch3. Furthermore, super enhancer interacted with the Tbx2 at the Lrch3 promoter locus, mediating the augmented transcription of circLrch3. CONCLUSION: Our findings clarify the role of a super enhancer-associated circLrch3 in the formation of R-loop with the host gene Lrch3 to modulate pyroptosis in PASMCs, ultimately promoting the development of PH.


Sujet(s)
Myocytes du muscle lisse , Artère pulmonaire , Pyroptose , ARN circulaire , Pyroptose/génétique , ARN circulaire/génétique , ARN circulaire/métabolisme , Animaux , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Myocytes du muscle lisse/métabolisme , Rats , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Hypoxie cellulaire/génétique , Muscles lisses vasculaires/métabolisme , Mâle , Hypertension pulmonaire/génétique , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Régulation de l'expression des gènes , Éléments activateurs (génétique)/génétique , Hypoxie/génétique , Hypoxie/métabolisme ,
16.
Genomics ; 116(3): 110840, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38580085

RÉSUMÉ

Conotruncal heart defects (CTD), subtypes of congenital heart disease, result from abnormal cardiac outflow tract development (OFT). FOXC1 and FOXC2 are closely related members of the forkhead transcription factor family and play essential roles in the development of OFT. We confirmed their expression pattern in mouse and human embryos, identifying four variants in FOXC1 and three in FOXC2 by screening these two genes in 605 patients with sporadic CTD. Western blot demonstrated expression levels, while Dual-luciferase reporter assay revealed affected transcriptional abilities for TBX1 enhancer in two FOXC1 variants and three FOXC2 variants. This might result from the altered DNA-binding abilities of mutant proteins. These results indicate that functionally impaired FOXC1 and FOXC2 variants may contribute to the occurrence of CTD.


Sujet(s)
Facteurs de transcription Forkhead , Cardiopathies congénitales , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Humains , Cardiopathies congénitales/génétique , Cardiopathies congénitales/métabolisme , Animaux , Souris , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme
17.
Circ Res ; 134(10): e112-e132, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38618720

RÉSUMÉ

BACKGROUND: The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease arising from defective morphogenesis of pharyngeal arch arteries (PAAs) and their derivatives. METHODS: Mouse genetics, lineage tracing, confocal microscopy, and quantitative image analyses were used to investigate mechanisms of PAA formation and repair. RESULTS: The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived endothelial cells (ECs) is regulated by VEGFR2 (vascular endothelial growth factor receptor 2) and Tbx1. Remarkably, when the SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated 3-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of 1 VEGFR2 allele (VEGFR2SHF-HET) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2SHF-KO) abolishes it. The decrease in SHF-derived ECs in VEGFR2SHF-HET and VEGFR2SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2SHF-KO mutants. Blocking the compensatory response in VEGFR2SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1+/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and congenital heart disease. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. CONCLUSIONS: Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling.


Sujet(s)
Aorte thoracique , Cellules endothéliales , Cardiopathies congénitales , Protéines à domaine boîte-T , Récepteur-2 au facteur croissance endothéliale vasculaire , Animaux , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , Souris , Aorte thoracique/embryologie , Aorte thoracique/métabolisme , Cardiopathies congénitales/génétique , Cardiopathies congénitales/métabolisme , Cardiopathies congénitales/anatomopathologie , Cardiopathies congénitales/embryologie , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Cellules endothéliales/métabolisme , Régulation de l'expression des gènes au cours du développement , Différenciation cellulaire , Souris de lignée C57BL
18.
Int J Mol Sci ; 25(6)2024 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-38542387

RÉSUMÉ

Mesenchymal-epithelial transition (MET) is a widely spread and evolutionarily conserved process across species during development. In Ciona embryogenesis, the notochord cells undergo the transition from the non-polarized mesenchymal state into the polarized endothelial-like state to initiate the lumen formation between adjacent cells. Based on previously screened MET-related transcription factors by ATAC-seq and Smart-Seq of notochord cells, Ciona robusta Snail (Ci-Snail) was selected for its high-level expression during this period. Our current knockout results demonstrated that Ci-Snail was required for notochord cell MET. Importantly, overexpression of the transcription factor Brachyury in notochord cells resulted in a similar phenotype with failure of lumen formation and MET. More interestingly, expression of Ci-Snail in the notochord cells at the late tailbud stage could partially rescue the MET defect caused by Brachyury-overexpression. These results indicated an inverse relationship between Ci-Snail and Brachyury during notochord cell MET, which was verified by RT-qPCR analysis. Moreover, the overexpression of Ci-Snail could significantly inhibit the transcription of Brachyury, and the CUT&Tag-qPCR analysis demonstrated that Ci-Snail is directly bound to the upstream region of Brachyury. In summary, we revealed that Ci-Snail promoted the notochord cell MET and was essential for lumen formation via transcriptionally repressing Brachyury.


Sujet(s)
Ciona intestinalis , Chorde , Animaux , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Facteurs de transcription/métabolisme , Protéines foetales/génétique , Protéines foetales/métabolisme , Ciona intestinalis/génétique , Régulation de l'expression des gènes au cours du développement
19.
Microbiol Spectr ; 12(5): e0364623, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38497717

RÉSUMÉ

Anti-interferon-γ autoantibody (AIGA) syndrome may be the basis of disseminated Talaromyces marneffei infection in human immunodeficiency virus (HIV)-negative adults. However, the pathogenesis of Th1 cell immunity in T. marneffei infection with AIGA syndrome is unknown. A multicenter study of HIV-negative individuals with T. marneffei infection was conducted between September 2018 and September 2020 in Guangdong and Guangxi, China. Patients were divided into AIGA-positive (AP) and AIGA-negative (AN) groups according to the AIGA titer and neutralizing activity. The relationship between AIGA syndrome and Th1 immune deficiency was investigated by using AP patient serum and purification of AIGA. Fifty-five HIV-negative adults with disseminated T. marneffei infection who were otherwise healthy were included. The prevalence of AIGA positivity was 83.6%. Based on their AIGA status, 46 and 9 patients were assigned to the AP and AN groups, respectively. The levels of Th1 cells, IFN-γ, and T-bet were higher in T. marneffei-infected patients than in healthy controls. However, the levels of CD4+ T-cell STAT-1 phosphorylation (pSTAT1) and Th1 cells were lower in the AP group than in the AN group. Both the serum of patients with AIGA syndrome and the AIGA purified from the serum of patients with AIGA syndrome could reduce CD4+ T-cell pSTAT1, Th1 cell differentiation and T-bet mRNA, and protein expression. The Th1 cell immune response plays a pivotal role in defense against T. marneffei infection in HIV-negative patients. Inhibition of the Th1 cell immune response may be an important pathological effect of AIGA syndrome.IMPORTANCEThe pathogenesis of Th1 cell immunity in Talaromyces marneffei infection with anti-interferon-γ autoantibody (AIGA) syndrome is unknown. This is an interesting study addressing an important knowledge gap regarding the pathogenesis of T. marneffei in non-HIV positive patients; in particular patients with AIGA. The finding of the Th1 cell immune response plays a pivotal role in defense against T. marneffei infection in HIV-negative patients, and inhibition of the Th1 cell immune response may be an important pathological effect of AIGA syndrome, which presented in this research could help bridge the current knowledge gap.


Sujet(s)
Autoanticorps , Interféron gamma , Mycoses , Talaromyces , Lymphocytes auxiliaires Th1 , Humains , Talaromyces/immunologie , Lymphocytes auxiliaires Th1/immunologie , Interféron gamma/immunologie , Autoanticorps/immunologie , Autoanticorps/sang , Mâle , Adulte , Femelle , Chine , Mycoses/immunologie , Mycoses/microbiologie , Adulte d'âge moyen , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/immunologie , Facteur de transcription STAT-1/immunologie , Facteur de transcription STAT-1/génétique
20.
Nucleic Acids Res ; 52(7): e40, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38499482

RÉSUMÉ

Genome-wide binding assays aspire to map the complete binding pattern of gene regulators. Common practice relies on replication-duplicates or triplicates-and high stringency statistics to favor false negatives over false positives. Here we show that duplicates and triplicates of CUT&RUN are not sufficient to discover the entire activity of transcriptional regulators. We introduce ICEBERG (Increased Capture of Enrichment By Exhaustive Replicate aGgregation), a pipeline that harnesses large numbers of CUT&RUN replicates to discover the full set of binding events and chart the line between false positives and false negatives. We employed ICEBERG to map the full set of H3K4me3-marked regions, the targets of the co-factor ß-catenin, and those of the transcription factor TBX3, in human colorectal cancer cells. The ICEBERG datasets allow benchmarking of individual replicates, comparing the performance of peak calling and replication approaches, and expose the arbitrary nature of strategies to identify reproducible peaks. Instead of a static view of genomic targets, ICEBERG establishes a spectrum of detection probabilities across the genome for a given factor, underlying the intrinsic dynamicity of its mechanism of action, and permitting to distinguish frequent from rare regulation events. Finally, ICEBERG discovered instances, undetectable with other approaches, that underlie novel mechanisms of colorectal cancer progression.


Sujet(s)
Logiciel , Transcription génétique , Humains , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Sites de fixation , Lignée cellulaire tumorale , Séquençage après immunoprécipitation de la chromatine , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Génome humain , Histone/métabolisme , Histone/génétique , Liaison aux protéines , Protéines à domaine boîte-T/génétique , Protéines à domaine boîte-T/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...