Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 856
Filtrer
1.
Respir Res ; 25(1): 313, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39154161

RÉSUMÉ

BACKGROUND: Due to a special hemodynamic feature, pulmonary vascular disease in pulmonary arterial hypertension associated with congenital heart disease (PAH-CHD) has two stages: reversible and irreversible. So far, the mechanism involved in the transition from reversible to irreversible stage is elusive. Moreover, no recognized and reliable assessments to distinguish these two stages are available. Furthermore, we found that compared with control and reversible PAH, thrombospondin-4 (THBS4) was significantly upregulated in irreversible group by bioinformatic analysis. Hence, we further verify and investigate the expression and role of THBS4 in PAH-CHD. METHODS: We established the monocrotaline plus aorto-cava shunt-induced (MCT-AV) rat model. We measured the expression of THBS4 in lung tissues from MCT-AV rats. Double immunofluorescence staining of lung tissue for THBS4 and α-SMA (biomarker of smooth muscle cells) or vWF (biomarker of endothelial cells) to identify the location of THBS4 in the pulmonary artery. Primary pulmonary artery smooth muscle cells (PASMCs) were cultivated, identified, and used in this study. THBS4 was inhibited and overexpressed by siRNA and plasmid, respectively, to explore the effect of THBS4 on phenotype transformation, proliferation, apoptosis, and migration of PASMCs. The effect of THBS4 on pulmonary vascular remodeling was evaluated in vivo by adeno-associated virus which suppressed THBS4 expression. Circulating level of THBS4 in patients with PAH-CHD was measured by ELISA. RESULTS: THBS4 was upregulated in the lung tissues of MCT-AV rats, and was further upregulated in severe pulmonary vascular lesions. And THBS4 was expressed mainly in PASMCs. When THBS4 was inhibited, contractile markers α-SMA and MYH11 were upregulated, while the proliferative marker PCNA was decreased, the endothelial-mensenchymal transition marker N-cad was downregulated, proapototic marker BAX was increased. Additionally, proliferation and migration of PASMCs was inhibited and apoptosis was increased. Conversely, THBS4 overexpression resulted in opposite effects. And the impact of THBS4 on PASMCs was probably achieved through the regulation of the PI3K/AKT pathway. THBS4 suppression attenuated pulmonary vascular remodeling. Furthermore, compared with patients with simple congenital heart disease and mild PAH-CHD, the circulating level of THBS4 was higher in patients with severe PAH-CHD. CONCLUSIONS: THBS4 is a promising biomarker to distinguish reversible from irreversible PAH-CHD before repairing the shunt. THBS4 is a potential treatment target in PAH-CHD, especially in irreversible stage.


Sujet(s)
Cardiopathies congénitales , Hypertension artérielle pulmonaire , Rat Sprague-Dawley , Thrombospondines , Animaux , Humains , Mâle , Rats , Cellules cultivées , Cardiopathies congénitales/métabolisme , Cardiopathies congénitales/complications , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Thrombospondines/métabolisme , Thrombospondines/biosynthèse , Thrombospondines/génétique
2.
Front Endocrinol (Lausanne) ; 15: 1416978, 2024.
Article de Anglais | MEDLINE | ID: mdl-39036049

RÉSUMÉ

Background: Endometriosis (EM) is a chronic painful condition that predominantly affects women of reproductive age. Currently, surgery or medication can only provide limited symptom relief. This study used a comprehensive genetic analytical approach to explore potential drug targets for EM in the plasma proteome. Methods: In this study, 2,923 plasma proteins were selected as exposure and EM as outcome for two-sample Mendelian randomization (MR) analyses. The plasma proteomic data were derived from the UK Biobank Pharmaceutical Proteomics Project (UKB-PPP), while the EM dataset from the FinnGen consortium R10 release data. Several sensitivity analyses were performed, including summary-data-based MR (SMR) analyses, heterogeneity in dependent instruments (HEIDI) test, reverse MR analyses, steiger detection test, and bayesian co-localization analyses. Furthermore, proteome-wide association study (PWAS) and single-cell transcriptomic analyses were also conducted to validate the findings. Results: Six significant (p < 3.06 × 10-5) plasma protein-EM pairs were identified by MR analyses. These included EPHB4 (OR = 1.40, 95% CI: 1.20 - 1.63), FSHB (OR = 3.91, 95% CI: 3.13 - 4.87), RSPO3 (OR = 1.60, 95% CI: 1.38 - 1.86), SEZ6L2 (OR = 1.44, 95% CI: 1.23 - 1.68) and WASHC3 (OR = 2.00, 95% CI: 1.54 - 2.59) were identified as risk factors, whereas KDR (OR = 0.80, 95% CI: 0.75 - 0.90) was found to be a protective factor. All six plasma proteins passed the SMR test (P < 8.33 × 10-3), but only four plasma proteins passed the HEIDI heterogeneity test (PHEIDI > 0.05), namely FSHB, RSPO3, SEZ6L2 and EPHB4. These four proteins showed strong evidence of co-localization (PPH4 > 0.7). In particular, RSPO3 and EPHB4 were replicated in the validated PWAS. Single-cell analyses revealed high expression of SEZ6L2 and EPHB4 in stromal and epithelial cells within EM lesions, while RSPO3 exhibited elevated expression in stromal cells and fibroblasts. Conclusion: Our study identified FSHB, RSPO3, SEZ6L2, and EPHB4 as potential drug targets for EM and highlighted the critical role of stromal and epithelial cells in disease development. These findings provide new insights into the diagnosis and treatment of EM.


Sujet(s)
Endométriose , Protéome , Protéomique , Humains , Femelle , Endométriose/sang , Endométriose/traitement médicamenteux , Endométriose/métabolisme , Protéome/métabolisme , Protéomique/méthodes , Protéines du sang/métabolisme , Adulte , Analyse de randomisation mendélienne , Marqueurs biologiques/sang , Étude d'association pangénomique , Thrombospondines/métabolisme , Thrombospondines/génétique
3.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-39062903

RÉSUMÉ

The differentiation between primary and secondary forms of membranous nephropathy (MN) is a cornerstone that is necessary for adequate decision making regarding the treatment options and behavior of each specific case. Kidney biopsy and antibody results can be controversial, and a unique biomarker has still not been found. BACKGROUND AND OBJECTIVES: We investigated the lack of mannose-binding lectin (MBL) deposition in patients with secondary MNs (sMNs) with the presence of IgG4 deposition in relation to the presence of MBL deposition in patients with primary MNs (pMNs). We also established a connection between the stage of MN and MBL deposition. MATERIALS AND METHODS: Materials from 72 renal biopsies with proven MN were used for immunohistochemistry staining (IHC) for the phospholipase A2 receptor (PLA2R), immunoglobulin subtype IgG4, and MBL. Patients were separated into one of the following three groups: primary MN (pMN), idiopathic MN (iMN), and secondary MN (sMN). Serum antibodies for PLA2R and thrombospondin type-I-domain-containing 7A (THSD7A) were also used for the precise evaluation of the type of MN, as well as for detecting positivity for PLA2R using IHC. Which stage of MN was present in relation to the deposition of MBL was evaluated. RESULTS: In total, 50 patients were positive for IgG4, 34 with pMN, 12 with iMN, and 4 with sMN. A total of 20 patients were positive for MBL, 14 with pMN and 6 with iMN; no MBL deposits were found in patients with sMN. MBL positivity was predominantly present in the first two stages of MN, with a gradual reduction in the later stages. CONCLUSIONS: The activation of the lectin-complement pathway occurs in the early stages of the disease and is associated with the deposition of IgG4; IgG4 deposition is present in sMN, but there is no MBL deposition. IgG4 cannot be used for the differentiation of primary from secondary MNs, but the lack of MBL can be used as a marker for sMN in the early stages of the disease.


Sujet(s)
Glomérulonéphrite extra-membraneuse , Immunoglobuline G , Lectine liant le mannose , Récepteurs à la phospholipase A2 , Humains , Glomérulonéphrite extra-membraneuse/métabolisme , Glomérulonéphrite extra-membraneuse/anatomopathologie , Glomérulonéphrite extra-membraneuse/immunologie , Glomérulonéphrite extra-membraneuse/diagnostic , Mâle , Femelle , Lectine liant le mannose/métabolisme , Adulte d'âge moyen , Immunoglobuline G/métabolisme , Immunoglobuline G/immunologie , Adulte , Récepteurs à la phospholipase A2/métabolisme , Récepteurs à la phospholipase A2/immunologie , Marqueurs biologiques , Sujet âgé , Thrombospondines/métabolisme , Rein/métabolisme , Rein/anatomopathologie , Biopsie
4.
Sci Rep ; 14(1): 13810, 2024 06 14.
Article de Anglais | MEDLINE | ID: mdl-38877292

RÉSUMÉ

Fibrosis is an important complication in inflammatory bowel diseases. Previous studies suggest an important role of matrix Gla protein (MGP) and thrombospondin 2 (THBS2) in fibrosis in various organs. Our aim was to analyse their expression together with regulatory miRNAs in submucosal and subserosal fibroblasts in ulcerative colitis (UC) and Crohn's disease (CD) using immunohistochemistry and qPCR. Digital pathology was used to compare collagen fibre characteristics of submucosal and subserosal fibrosis. Immunohistochemistry showed expression of MGP, but not THBS2 in submucosa in UC and CD. In the subserosa, there was strong staining for both proteins in CD but not in UC. qPCR showed significant upregulation of THBS2 and MGP genes in CD subserosa compared to the submucosa. Digital pathology analysis revealed higher proportion of larger and thicker fibres that were more tortuous and reticulated in subserosal fibrosis compared to submucosal fibrosis. These results suggest distinct fibroblast populations in fibrostenosing CD, and are further supported by image analysis showing significant differences in the morphology and architecture of collagen fibres in submucosal fibrosis in comparison to subserosal fibrosis. Our study is the first to describe differences in submucosal and subserosal fibroblast populations, contributing to understanding of the pathogenesis of fibrostenosis in CD.


Sujet(s)
Protéines de liaison au calcium , Maladie de Crohn , Protéines de la matrice extracellulaire , Fibroblastes , Fibrose , , Thrombospondines , Maladie de Crohn/anatomopathologie , Maladie de Crohn/métabolisme , Humains , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Protéines de la matrice extracellulaire/métabolisme , Protéines de liaison au calcium/métabolisme , Protéines de liaison au calcium/génétique , Thrombospondines/métabolisme , Thrombospondines/génétique , Mâle , Femelle , Adulte , Adulte d'âge moyen , Rectocolite hémorragique/anatomopathologie , Rectocolite hémorragique/métabolisme , microARN/génétique , microARN/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/métabolisme , Sujet âgé , Immunohistochimie
5.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892120

RÉSUMÉ

Following the discovery of podocyte phospholipase A2 receptor and thrombospondin type-1 domain-containing 7A, various potential target antigens for membranous nephropathy (MN) have been reported one after another. MN target antigens have now been identified in a significant proportion of patients, and a new classification framework classifies patients with MN based on the detected antigen and associated disease phenotype. A serology-based approach that does not require a histological diagnosis for patients suspected of having MN has also been proposed. However, there have been cases in which dual positivity for MN antigens and/or corresponding antibodies has been shown. Importantly, some of them showed a transition of the affected patient's immune responses to MN antigens, suggesting that serological diagnosis changes depending on the timing of the analysis. In this review, we provide detailed information on these cases and present an overview of our recent understanding of their putative mechanisms involved in these cases. Greater awareness is required to adequately recognize and develop appropriate therapeutic strategies for this condition.


Sujet(s)
Glomérulonéphrite extra-membraneuse , Glomérulonéphrite extra-membraneuse/immunologie , Glomérulonéphrite extra-membraneuse/diagnostic , Glomérulonéphrite extra-membraneuse/sang , Humains , Récepteurs à la phospholipase A2/immunologie , Récepteurs à la phospholipase A2/métabolisme , Autoantigènes/immunologie , Prévalence , Podocytes/métabolisme , Podocytes/immunologie , Podocytes/anatomopathologie , Autoanticorps/immunologie , Autoanticorps/sang , Thrombospondines/immunologie , Thrombospondines/métabolisme
7.
Sci Rep ; 14(1): 14757, 2024 06 26.
Article de Anglais | MEDLINE | ID: mdl-38926599

RÉSUMÉ

Muscular dystrophy is a group of genetic disorders that lead to muscle wasting and loss of muscle function. Identifying genetic modifiers that alleviate symptoms or enhance the severity of a primary disease helps to understand mechanisms behind disease pathology and facilitates discovery of molecular targets for therapy. Several muscular dystrophies are caused by genetic defects in the components of the dystrophin-glycoprotein adhesion complex (DGC). Thrombospondin-4 overexpression has been shown to mitigate dystrophic disease in mouse models for Duchenne muscular dystrophy (dystrophin deficiency) and limb-girdle muscular dystrophy type 2F (LGMD2F, δ-sarcoglycan deficiency), while deletion of the thrombospondin-4 gene exacerbated the diseases. Hence, thrombospondin-4 has been considered a candidate molecule for therapy of muscular dystrophies involving the DGC. We have investigated whether thrombospondin-4 could act as a genetic modifier for other DGC-associated diseases: limb-girdle muscular dystrophy type 2E (LGMD2E, ß-sarcoglycan deficiency) and laminin α2 chain-deficient muscular dystrophy (LAMA2-RD). Deletion of the thrombospondin-4 gene in mouse models for LGMD2E and LAMA2-RD, respectively, did not result in worsening of the dystrophic phenotype. Loss of thrombospondin-4 did not enhance sarcolemma damage and did not impair trafficking of transmembrane receptors integrin α7ß1 and dystroglycan in double knockout muscles. Our results suggest that thrombospondin-4 might not be a relevant therapeutic target for all muscular dystrophies involving the DGC. This data also demonstrates that molecular pathology between very similar diseases like LGMD2E and 2F can differ significantly.


Sujet(s)
Laminine , Souris knockout , Sarcoglycanes , Thrombospondines , Animaux , Laminine/métabolisme , Laminine/génétique , Laminine/déficit , Sarcoglycanes/génétique , Sarcoglycanes/déficit , Sarcoglycanes/métabolisme , Souris , Thrombospondines/génétique , Thrombospondines/métabolisme , Thrombospondines/déficit , Modèles animaux de maladie humaine , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Délétion de gène , Dystrophies musculaires/génétique , Dystrophies musculaires/métabolisme , Dystrophies musculaires/anatomopathologie , Dystrophie musculaire de l'animal/génétique , Dystrophie musculaire de l'animal/métabolisme , Dystrophie musculaire de l'animal/anatomopathologie
8.
Sci Adv ; 10(20): eadm9326, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758792

RÉSUMÉ

Intellectual disability (ID) affects ~2% of the population and ID-associated genes are enriched for epigenetic factors, including those encoding the largest family of histone lysine acetyltransferases (KAT5-KAT8). Among them is KAT6A, whose mutations cause KAT6A syndrome, with ID as a common clinical feature. However, the underlying molecular mechanism remains unknown. Here, we find that KAT6A deficiency impairs synaptic structure and plasticity in hippocampal CA3, but not in CA1 region, resulting in memory deficits in mice. We further identify a CA3-enriched gene Rspo2, encoding Wnt activator R-spondin 2, as a key transcriptional target of KAT6A. Deletion of Rspo2 in excitatory neurons impairs memory formation, and restoring RSPO2 expression in CA3 neurons rescues the deficits in Wnt signaling and learning-associated behaviors in Kat6a mutant mice. Collectively, our results demonstrate that KAT6A-RSPO2-Wnt signaling plays a critical role in regulating hippocampal CA3 synaptic plasticity and cognitive function, providing potential therapeutic targets for KAT6A syndrome and related neurodevelopmental diseases.


Sujet(s)
Cognition , Histone acetyltransferases , Thrombospondines , Voie de signalisation Wnt , Animaux , Souris , Région CA3 de l'hippocampe/métabolisme , Région CA3 de l'hippocampe/anatomopathologie , Histone acetyltransferases/déficit , Histone acetyltransferases/génétique , Histone acetyltransferases/métabolisme , Souris knockout , Plasticité neuronale , Thrombospondines/génétique , Thrombospondines/métabolisme
10.
Int J Biol Macromol ; 269(Pt 2): 131805, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38677673

RÉSUMÉ

Endothelial repair is essential for restoring tissue fluid homeostasis following lung injury. R-spondin3 (RSPO3), a secreted protein mainly produced by endothelial cells (ECs), has shown its protective effect on endothelium. However, the specific mechanisms remain unknown. To explore whether and how RSPO3 regulates endothelial regeneration after inflammatory vascular injury, the role of RSPO3 in sepsis-induced pulmonary endothelial injury was investigated in EC-specific RSPO3 knockdown, inducible EC-specific RSPO3 deletion mice, EC-specific RSPO3 overexpression mice, systemic RSPO3-administration mice, in isolated mouse lung vascular endothelial cells (MLVECs), and in plasma from septic patients. Here we show that plasma RSPO3 levels are decreased in septic patients and correlated with endothelial injury markers and PaO2/FiO2 index. Both pulmonary EC-specific knockdown of RSPO3 and inducible EC-specific RSPO3 deletion inhibit pulmonary ECs proliferation and exacerbate ECs injury, whereas intra-pulmonary EC-specific RSPO3 overexpression promotes endothelial recovery and attenuates ECs injury during endotoxemia. We show that RSPO3 mediates pulmonary endothelial regeneration by a LGR4-dependent manner. Except for ß-catenin, integrin-linked kinase (ILK)/Akt is also identified as a novel downstream effector of RSPO3/LGR4 signaling. These results conclude that EC-derived RSPO3 mediates pulmonary endothelial regeneration by LGR4-dependent activation of ß-catenin and ILK signaling pathways after inflammatory vascular injury.


Sujet(s)
Cellules endothéliales , Poumon , Protein-Serine-Threonine Kinases , Récepteurs couplés aux protéines G , Régénération , Transduction du signal , Thrombospondines , bêta-Caténine , Animaux , Thrombospondines/métabolisme , Thrombospondines/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Souris , Humains , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Cellules endothéliales/métabolisme , Poumon/anatomopathologie , Poumon/métabolisme , Lésions du système vasculaire/métabolisme , Lésions du système vasculaire/génétique , Lésions du système vasculaire/anatomopathologie , Prolifération cellulaire , Mâle , Sepsie/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie
11.
J Biol Chem ; 300(5): 107284, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38614208

RÉSUMÉ

Receptor-mediated cellular uptake of specific ligands constitutes an important step in the dynamic regulation of individual protein levels in extracellular fluids. With a focus on the inflammatory lung, we here performed a proteomics-based search for novel ligands regulated by the mannose receptor (MR), a macrophage-expressed endocytic receptor. WT and MR-deficient mice were exposed to lipopolysaccharide, after which the protein content in their lung epithelial lining fluid was compared by tandem mass tag-based mass spectrometry. More than 1200 proteins were identified in the epithelial lining fluid using this unbiased approach, but only six showed a statistically different abundance. Among these, an unexpected potential new ligand, thrombospondin-4 (TSP-4), displayed a striking 17-fold increased abundance in the MR-deficient mice. Experiments using exogenous addition of TSP-4 to MR-transfected CHO cells or MR-positive alveolar macrophages confirmed that TSP-4 is a ligand for MR-dependent endocytosis. Similar studies revealed that the molecular interaction with TSP-4 depends on both the lectin activity and the fibronectin type-II domain of MR and that a closely related member of the TSP family, TSP-5, is also efficiently internalized by the receptor. This was unlike the other members of this protein family, including TSPs -1 and -2, which are ligands for a close MR homologue known as urokinase plasminogen activator receptor-associated protein. Our study shows that MR takes part in the regulation of TSP-4, an important inflammatory component in the injured lung, and that two closely related endocytic receptors, expressed on different cell types, undertake the selective endocytosis of distinct members of the TSP family.


Sujet(s)
Lectines de type C , Lésion pulmonaire , Récepteur du mannose , Lectines liant le mannose , Protéomique , Récepteurs de surface cellulaire , Thrombospondines , Animaux , Souris , Cellules CHO , Cricetulus , Endocytose , Lectines de type C/métabolisme , Lectines de type C/génétique , Ligands , Lipopolysaccharides/toxicité , Poumon/métabolisme , Poumon/anatomopathologie , Lésion pulmonaire/métabolisme , Lésion pulmonaire/anatomopathologie , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/anatomopathologie , Lectines liant le mannose/métabolisme , Lectines liant le mannose/génétique , Souris knockout , Protéomique/méthodes , Récepteurs de surface cellulaire/métabolisme , Récepteurs de surface cellulaire/génétique , Thrombospondines/métabolisme , Thrombospondines/génétique
12.
Exp Neurol ; 376: 114756, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38508482

RÉSUMÉ

Overexpression of the Ube3a gene and the resulting increase in Ube3a protein are linked to autism spectrum disorder (ASD). However, the cellular and molecular processes underlying Ube3a-dependent ASD remain unclear. Using both male and female mice, we find that neurons in the somatosensory cortex of the Ube3a 2× Tg ASD mouse model display reduced dendritic spine density and increased immature filopodia density. Importantly, the increased gene dosage of Ube3a in astrocytes alone is sufficient to confer alterations in neurons as immature dendritic protrusions, as observed in primary hippocampal neuron cultures. We show that Ube3a overexpression in astrocytes leads to a loss of astrocyte-derived spinogenic protein, thrombospondin-2 (TSP2), due to a suppression of TSP2 gene transcription. By neonatal intraventricular injection of astrocyte-specific virus, we demonstrate that Ube3a overexpression in astrocytes in vivo results in a reduction in dendritic spine maturation in prelimbic cortical neurons, accompanied with autistic-like behaviors in mice. These findings reveal an astrocytic dominance in initiating ASD pathobiology at the neuronal and behavior levels. SIGNIFICANCE STATEMENT: Increased gene dosage of Ube3a is tied to autism spectrum disorders (ASDs), yet cellular and molecular alterations underlying autistic phenotypes remain unclear. We show that Ube3a overexpression leads to impaired dendritic spine maturation, resulting in reduced spine density and increased filopodia density. We find that dysregulation of spine development is not neuron autonomous, rather, it is mediated by an astrocytic mechanism. Increased gene dosage of Ube3a in astrocytes leads to reduced production of the spinogenic glycoprotein thrombospondin-2 (TSP2), leading to abnormalities in spines. Astrocyte-specific Ube3a overexpression in the brain in vivo confers dysregulated spine maturation concomitant with autistic-like behaviors in mice. These findings indicate the importance of astrocytes in aberrant neurodevelopment and brain function in Ube3a-depdendent ASD.


Sujet(s)
Trouble du spectre autistique , Épines dendritiques , Névroglie , Ubiquitin-protein ligases , Animaux , Souris , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/génétique , Trouble du spectre autistique/anatomopathologie , Cellules cultivées , Épines dendritiques/anatomopathologie , Épines dendritiques/métabolisme , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Souris de lignée C57BL , Souris transgéniques , Neurogenèse/physiologie , Névroglie/métabolisme , Névroglie/anatomopathologie , Neurones/métabolisme , Neurones/anatomopathologie , Cortex somatosensoriel/métabolisme , Cortex somatosensoriel/anatomopathologie , Thrombospondines/métabolisme , Thrombospondines/génétique , Thrombospondines/biosynthèse , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme
13.
Eur J Hum Genet ; 32(5): 550-557, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38433265

RÉSUMÉ

Ehlers-Danlos syndromes (EDS) are a group of connective tissue disorders caused by mutations in collagen and collagen-interacting genes. We delineate a novel form of EDS with vascular features through clinical and histopathological phenotyping and genetic studies of a three-generation pedigree, displaying an apparently autosomal dominant phenotype of joint hypermobility and frequent joint dislocations, atrophic scarring, prolonged bleeding time and age-related aortic dilatation and rupture. Coagulation tests as well as platelet counts and function were normal. Reticular dermis displayed highly disorganized collagen fibers and transmission electron microscopy (TEM) revealed abnormally shaped fibroblasts and endothelial cells, with high amount and irregular shape of extracellular matrix (ECM) substance, especially near blood vessels. Genetic analysis unraveled a heterozygous mutation in THBS2 (NM_003247.5:c.2686T>C, p.Cys896Arg). We generated CRISPR/Cas9 knock-in (KI) mice, bearing the heterozygous human mutation in the mouse ortholog. The KI mice demonstrated phenotypic traits correlating with those observed in the human subjects, as evidenced by morphologic, histologic, and TEM analyses, in conjunction with bleeding time assays. Our findings delineate a novel form of human EDS with classical-like elements combined with vascular features, caused by a heterozygous THBS2 missense mutation. We further demonstrate a similar phenotype in heterozygous THBS2Cys896Arg KI mice, in line with previous studies in Thbs2 homozygous null-mutant mice. Notably, THBS2 encodes Thrombospondin-2, a secreted homotrimeric matricellular protein that directly binds the ECM-shaping Matrix Metalloproteinase 2 (MMP2), mediating its clearance. THBS2 loss-of-function attenuates MMP2 clearance, enhancing MMP2-mediated proteoglycan cleavage, causing ECM abnormalities similar to those seen in the human and mouse disease we describe.


Sujet(s)
Syndrome d'Ehlers-Danlos , Hétérozygote , Thrombospondines , Syndrome d'Ehlers-Danlos/génétique , Syndrome d'Ehlers-Danlos/anatomopathologie , Syndrome d'Ehlers-Danlos/métabolisme , Animaux , Thrombospondines/génétique , Thrombospondines/métabolisme , Humains , Souris , Mâle , Femelle , Adulte , Phénotype , Pedigree , Adulte d'âge moyen , Mutation faux-sens
14.
Front Cell Infect Microbiol ; 14: 1354880, 2024.
Article de Anglais | MEDLINE | ID: mdl-38465236

RÉSUMÉ

Plasmodium vivax, the most widespread human malaria parasite, and P. knowlesi, an emerging Plasmodium that infects humans, are the phylogenetically closest malarial species that infect humans, which may induce cross-species reactivity across most co-endemic areas in Southeast Asia. The thrombospondin-related anonymous protein (TRAP) family is indispensable for motility and host cell invasion in the growth and development of Plasmodium parasites. The merozoite-specific TRAP (MTRAP), expressed in blood-stage merozoites, is supposed to be essential for human erythrocyte invasion. We aimed to characterize MTRAPs in blood-stage P. vivax and P. knowlesi parasites and ascertain their cross-species immunoreactivity. Recombinant P. vivax and P. knowlesi MTRAPs of full-length ectodomains were expressed in a mammalian expression system. The MTRAP-specific immunoglobulin G, obtained from immune animals, was used in an immunofluorescence assay for subcellular localization and invasion inhibitory activity in blood-stage parasites was determined. The cross-species humoral immune responses were analyzed in the sera of patients with P. vivax or P. knowlesi infections. The MTRAPs of P. vivax (PvMTRAP) and P. knowlesi (PkMTRAP) were localized on the rhoptry body of merozoites in blood-stage parasites. Both anti-PvMTRAP and anti-PkMTRAP antibodies inhibited erythrocyte invasion of blood-stage P. knowlesi parasites. The humoral immune response to PvMTRAP showed high immunogenicity, longevity, and cross-species immunoreactivity with P. knowlesi. MTRAPs are promising candidates for development of vaccines and therapeutics against vivax and knowlesi malaria.


Sujet(s)
Paludisme à Plasmodium vivax , Paludisme , Parasites , Plasmodium , Animaux , Humains , Plasmodium vivax/génétique , Parasites/métabolisme , Mérozoïtes , Thrombospondines/métabolisme , Plasmodium/métabolisme , Paludisme/parasitologie , Paludisme à Plasmodium vivax/parasitologie , Protéines de protozoaire/métabolisme , Mammifères/métabolisme
15.
Biol Reprod ; 110(5): 985-999, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38376238

RÉSUMÉ

Sry on the Y-chromosome upregulates Sox9, which in turn upregulates a set of genes such as Fgf9 to initiate testicular differentiation in the XY gonad. In the absence of Sry expression, genes such as Rspo1, Foxl2, and Runx1 support ovarian differentiation in the XX gonad. These two pathways antagonize each other to ensure the development of only one gonadal sex in normal development. In the B6.YTIR mouse, carrying the YTIR-chromosome on the B6 genetic background, Sry is expressed in a comparable manner with that in the B6.XY mouse, yet, only ovaries or ovotestes develop. We asked how testicular and ovarian differentiation pathways interact to determine the gonadal sex in the B6.YTIR mouse. Our results showed that (1) transcript levels of Sox9 were much lower than in B6.XY gonads while those of Rspo1 and Runx1 were as high as B6.XX gonads at 11.5 and 12.5 days postcoitum. (2) FOXL2-positive cells appeared in mosaic with SOX9-positive cells at 12.5 days postcoitum. (3) SOX9-positive cells formed testis cords in the central area while those disappeared to leave only FOXL2-positive cells in the poles or the entire area at 13.5 days postcoitum. (4) No difference was found at transcript levels of all genes between the left and right gonads up to 12.5 days postcoitum, although ovotestes developed much more frequently on the left than the right at 13.5 days postcoitum. These results suggest that inefficient Sox9 upregulation and the absence of Rspo1 repression prevent testicular differentiation in the B6.YTIR gonad.


Sujet(s)
Facteur de transcription SOX-9 , Processus de détermination du sexe , Testicule , Thrombospondines , Régulation positive , Animaux , Facteur de transcription SOX-9/génétique , Facteur de transcription SOX-9/métabolisme , Mâle , Femelle , Souris , Thrombospondines/génétique , Thrombospondines/métabolisme , Processus de détermination du sexe/génétique , Processus de détermination du sexe/physiologie , Testicule/métabolisme , Gonades/métabolisme , Ovaire/métabolisme , Protéine L2 à motif en tête de fourche/génétique , Protéine L2 à motif en tête de fourche/métabolisme , Sous-unité alpha 2 du facteur CBF/génétique , Sous-unité alpha 2 du facteur CBF/métabolisme , Régulation de l'expression des gènes au cours du développement , Différenciation sexuelle/génétique , Souris de lignée C57BL
16.
Eur J Cell Biol ; 103(2): 151395, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38340499

RÉSUMÉ

Pain is the most common symptom for which patients seek medical attention. Existing treatments for pain control are largely ineffective due to the lack of an accurate way to objectively measure pain intensity and a poor understanding of the etiology of pain. Thrombospondin 4(TSP4), a member of the thrombospondin gene family, is expressed in neurons and astrocytes and induces pain by interacting with the calcium channel alpha-2-delta-1 subunit (Cavα2δ1). In the present study we show that TSP4 expression level correlates positively with pain intensity, suggesting that TSP4 could be a novel candidate of pain indicator. Using RNAi-lentivirus (RNAi-LV) to knock down TSP4 both in vivo and in vitro, together with electrophysiological experiments involving paired patch-clamp recordings of evoked action potentials and post-synaptic currents in cultured neurons, we found that TSP4 contributes to the development of bone cancer pain, neuropathic pain, and inflammatory pain. This effect is mediated by regulation of neuron excitability via inhibition of synapsin I (Syn I) and modulation of excitatory and inhibitory presynaptic transmission via regulation of vesicular glutamate transporter 2(Vglut2), vesicular GABA transporter (VGAT), and glutamate decarboxylase (GAD) expression. The present study provides a replicable, predictive, valid indicator of pain and demonstrated the underlying molecular and electrophysiological mechanisms by which TSP4 contributes to pain.


Sujet(s)
Thrombospondines , Animaux , Thrombospondines/métabolisme , Thrombospondines/génétique , Mâle , Douleur/métabolisme , Neurones/métabolisme , Souris , Humains , Femelle , Tumeurs osseuses/métabolisme , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie
17.
Int J Mol Sci ; 25(4)2024 Feb 10.
Article de Anglais | MEDLINE | ID: mdl-38396816

RÉSUMÉ

Focal adhesions (FAs) play a crucial role in cell spreading and adhesion, and their autophagic degradation is an emerging area of interest. This study investigates the role of Thrombospondin Type 1 Domain-Containing Protein 1 (THSD1) in regulating autophagy and FA stability in brain endothelial cells, shedding light on its potential implications for cerebrovascular diseases. Our research reveals a physical interaction between THSD1 and FAs. Depletion of THSD1 significantly reduces FA numbers, impairing cell spreading and adhesion. The loss of THSD1 also induces autophagy independently of changes in mTOR and AMPK activation, implying that THSD1 primarily governs FA dynamics rather than serving as a global regulator of nutrient and energy status. Mechanistically, THSD1 negatively regulates Beclin 1, a central autophagy regulator, at FAs through interactions with focal adhesion kinase (FAK). THSD1 inactivation diminishes FAK activity and relieves its inhibitory phosphorylation on Beclin 1. This, in turn, promotes the complex formation between Beclin 1 and ATG14, a critical event for the activation of the autophagy cascade. In summary, our findings identify THSD1 as a novel regulator of autophagy that degrades FAs in brain endothelial cells. This underscores the distinctive nature of THSD1-mediated, cargo-directed autophagy and its potential relevance to vascular diseases due to the loss of endothelial FAs. Investigating the underlying mechanisms of THSD1-mediated pathways holds promise for discovering novel therapeutic targets in vascular diseases.


Sujet(s)
Contacts focaux , Thrombospondines , Maladies vasculaires , Humains , Autophagie , Bécline-1/métabolisme , Cellules endothéliales/métabolisme , Focal adhesion protein-tyrosine kinases/métabolisme , Contacts focaux/métabolisme , Phosphorylation , Maladies vasculaires/métabolisme , Thrombospondines/métabolisme
18.
Matrix Biol ; 127: 8-22, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38281553

RÉSUMÉ

Lumbar spinal canal stenosis is primarily caused by ligamentum flavum hypertrophy (LFH), which is a significant pathological factor. Nevertheless, the precise molecular basis for the development of LFH remains uncertain. The current investigation observed a notable increase in thrombospondin-1 (THBS1) expression in LFH through proteomics analysis and single-cell RNA-sequencing analysis of clinical ligamentum flavum specimens. In laboratory experiments, it was demonstrated that THBS1 triggered the activation of Smad3 signaling induced by transforming growth factor ß1 (TGFß1), leading to the subsequent enhancement of COL1A2 and α-SMA, which are fibrosis markers. Furthermore, experiments conducted on a bipedal standing mouse model revealed that THBS1 played a crucial role in the development of LFH. Sestrin2 (SESN2) acted as a stress-responsive protein that suppressed the expression of THBS1, thus averting the progression of fibrosis in ligamentum flavum (LF) cells. To summarize, these results indicate that mechanical overloading causes an increase in THBS1 production, which triggers the TGFß1/Smad3 signaling pathway and ultimately results in the development of LFH. Targeting the suppression of THBS1 expression may present a novel approach for the treatment of LFH.


Sujet(s)
Ligament jaune , Protéine Smad-3 , Thrombospondines , Facteur de croissance transformant bêta-1 , Animaux , Souris , Fibrose , Hypertrophie/métabolisme , Ligament jaune/métabolisme , Ligament jaune/anatomopathologie , Transduction du signal , Contrainte mécanique , Thrombospondines/génétique , Thrombospondines/métabolisme , Facteur de croissance transformant bêta-1/génétique , Facteur de croissance transformant bêta-1/métabolisme , Protéine Smad-3/génétique , Protéine Smad-3/métabolisme
19.
Cell Commun Signal ; 22(1): 14, 2024 01 05.
Article de Anglais | MEDLINE | ID: mdl-38183076

RÉSUMÉ

R-spondins (RSPOs) are secreted signaling molecules that potentiate the Wnt/ß-catenin pathway by cooperating with Wnt ligands. RSPO1 is crucial in tissue development and tissue homeostasis. However, the molecular mechanism by which RSPOs activate Wnt/ß-catenin signaling remains elusive. In this study, we found that RSPOs could mediate the degradation of Axin through the ubiquitin-proteasome pathway. The results of Co-IP showed that the recombinant RSPO1 protein promoted the interaction between Axin1 and CK1ε. Either knockout of the CK1ε gene or treatment with the CK1δ/CK1ε inhibitor SR3029 caused an increase in Axin1 protein levels and attenuated RSPO1-induced degradation of the Axin1 protein. Moreover, we observed an increase in the number of associations of LRP6 with CK1ε and Axin1 following RSPO1 stimulation. Overexpression of LRP6 further potentiated Axin1 degradation mediated by RSPO1 or CK1ε. In addition, recombinant RSPO1 and Wnt3A proteins synergistically downregulated the protein expression of Axin1 and enhanced the transcriptional activity of the SuperTOPFlash reporter. Taken together, these results uncover the novel mechanism by which RSPOs activate Wnt/ß-catenin signaling through LRP6/CK1ε-mediated degradation of Axin.


Sujet(s)
Axine , Thrombospondines , Voie de signalisation Wnt , bêta-Caténine , Transport biologique , Protéine Wnt3A , Humains , Thrombospondines/métabolisme
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167026, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38237742

RÉSUMÉ

Coal mining carries inherent risks of catastrophic gas explosions capable of inflicting severe lung injury. Using complementary in vivo and in vitro models, we explored mechanisms underlying alveolar epithelial damage and repair following a gas explosion in this study. In a rat model, the gas explosion was demonstrated to trigger inflammation and injury within the alveolar epithelium. The following scRNA-sequencing revealed that alveolar epithelial cells exhibited the most profound transcriptomic changes after gas explosion compared to other pulmonary cell types. In the L2 alveolar epithelial cells, the blast was found to cause autophagic flux by inducing autophagosome formation, LC3 lipidation, and p62 degradation. Transcriptomic profiling of the L2 cells identified PI3K-Akt and p53 pathways as critical modulators governing autophagic and oxidative stress responses to blast damage. Notably, Thrombospondin-1 (Thbs1) was determined for the first time as a pivotal node interconnecting these two pathways. The findings of this study illuminate intricate mechanisms of alveolar epithelial injury and recovery after blast trauma, highlighting autophagic and oxidative stress responses mediated by Thbs1-associated PI3K-Akt and p53 pathways as high-value therapeutic targets, and strategic modulation of these pathways in future studies may mitigate lung damage by reducing oxidative stress while engaging endogenous tissue repair processes like autophagy.


Sujet(s)
Lésion pulmonaire , Rats , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Stress oxydatif , Autophagie , Thrombospondines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE