Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48.894
Filtrer
1.
PLoS One ; 19(7): e0306248, 2024.
Article de Anglais | MEDLINE | ID: mdl-38950058

RÉSUMÉ

Diabetic foot ulcers (DFUs) pose a significant challenge in diabetes care. Yet, a comprehensive understanding of the underlying biological disparities between healing and non-healing DFUs remains elusive. We conducted bioinformatics analysis of publicly available transcriptome sequencing data in an attempt to elucidate these differences. Our analysis encompassed differential analysis to unveil shifts in cell composition and gene expression profiles between non-healing and healing DFUs. Cell communication alterations were explored employing the Cellchat R package. Pseudotime analysis and cytoTRACE allowed us to dissect the heterogeneity within fibroblast subpopulations. Our findings unveiled disruptions in various cell types, localized low-grade inflammation, compromised systemic antigen processing and presentation, and extensive extracellular matrix signaling disarray in non-healing DFU patients. Some of these anomalies partially reverted in healing DFUs, particularly within the abnormal ECM-receptor signaling pathway. Furthermore, we distinguished distinct fibroblast subpopulations in non-healing and healing DFUs, each with unique biological functions. Healing-associated fibroblasts exhibited heightened extracellular matrix (ECM) remodeling and a robust wound healing response, while non-healing-associated fibroblasts showed signs of cellular senescence and complement activation, among other characteristics. This analysis offers profound insights into the wound healing microenvironment, identifies pivotal cell types for DFU healing promotion, and reveals potential therapeutic targets for DFU management.


Sujet(s)
Pied diabétique , Fibroblastes , Analyse sur cellule unique , Transcriptome , Cicatrisation de plaie , Pied diabétique/génétique , Pied diabétique/anatomopathologie , Pied diabétique/métabolisme , Humains , Cicatrisation de plaie/génétique , Analyse sur cellule unique/méthodes , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Matrice extracellulaire/métabolisme , Matrice extracellulaire/génétique , Analyse de profil d'expression de gènes , Transduction du signal/génétique
2.
Chin J Dent Res ; 27(2): 121-131, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38953477

RÉSUMÉ

As the biological mechanisms of orthodontic tooth movement have been explored further, scholars have gradually focused on the remodelling mechanism of the extracellular matrix (ECM) in the periodontal ligament (PDL). The ECM of the PDL consists of various types of collagens and other glycoproteins. The specific process and mechanism of ECM remodelling during orthodontic tooth movement remains unclear. Collagen I and III, which constitute major components of the PDL, are upregulated under orthodontic force. The changes in the contents of ECM proteins also depend on the expression of ECM-related enzymes, which organise new collagen fibre networks to adapt to changes in tooth position. The matrix metalloproteinase family is the main enzyme that participates in collagen hydrolysis and renewal and changes its expression under orthodontic force. Moreover, ECM adhesion molecules, such as integrins, are also regulated by orthodontic force and participate in the dynamic reaction of cell adhesion and separation with the ECM. This article reviews the changes in ECM components, related enzymes and adhesion molecules in the PDL under orthodontic force to lay the foundation for the exploration of the regulatory mechanism of ECM remodelling during orthodontic tooth movement.


Sujet(s)
Matrice extracellulaire , Desmodonte , Mouvement dentaire , Matrice extracellulaire/métabolisme , Humains , Mouvement dentaire/méthodes , Desmodonte/cytologie , Parodonte/métabolisme , Matrix metalloproteinases/métabolisme , Intégrines/métabolisme , Collagène/métabolisme
3.
Proc Natl Acad Sci U S A ; 121(28): e2404210121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38954541

RÉSUMÉ

Mesenchymal stem cells (MSCs) are essential in regenerative medicine. However, conventional expansion and harvesting methods often fail to maintain the essential extracellular matrix (ECM) components, which are crucial for their functionality and efficacy in therapeutic applications. Here, we introduce a bone marrow-inspired macroporous hydrogel designed for the large-scale production of MSC-ECM spheroids. Through a soft-templating approach leveraging liquid-liquid phase separation, we engineer macroporous hydrogels with customizable features, including pore size, stiffness, bioactive ligand distribution, and enzyme-responsive degradability. These tailored environments are conducive to optimal MSC proliferation and ease of harvesting. We find that soft hydrogels enhance mechanotransduction in MSCs, establishing a standard for hydrogel-based 3D cell culture. Within these hydrogels, MSCs exist as both cohesive spheroids, preserving their innate vitality, and as migrating entities that actively secrete functional ECM proteins. Additionally, we also introduce a gentle, enzymatic harvesting method that breaks down the hydrogels, allowing MSCs and secreted ECM to naturally form MSC-ECM spheroids. These spheroids display heightened stemness and differentiation capacity, mirroring the benefits of a native ECM milieu. Our research underscores the significance of sophisticated materials design in nurturing distinct MSC subpopulations, facilitating the generation of MSC-ECM spheroids with enhanced therapeutic potential.


Sujet(s)
Matrice extracellulaire , Hydrogels , Cellules souches mésenchymateuses , Sphéroïdes de cellules , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Hydrogels/composition chimique , Matrice extracellulaire/métabolisme , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Humains , Différenciation cellulaire , Techniques de culture cellulaire/méthodes , Prolifération cellulaire , Porosité , Mécanotransduction cellulaire/physiologie , Cellules cultivées
4.
Front Immunol ; 15: 1379570, 2024.
Article de Anglais | MEDLINE | ID: mdl-38957465

RÉSUMÉ

There is a reciprocal relationship between extracellular matrix (ECM) remodelling and inflammation that could be operating in the progression of severe COVID-19. To explore the immune-driven ECM remodelling in COVID-19, we in this explorative study analysed these interactions in hospitalised COVID-19 patients. RNA sequencing and flow analysis were performed on peripheral blood mononuclear cells. Inflammatory mediators in plasma were measured by ELISA and MSD, and clinical information from hospitalised COVID-19 patients (N=15) at admission was included in the analysis. Further, we reanalysed two publicly available datasets: (1) lung tissue RNA-sequencing dataset (N=5) and (2) proteomics dataset from PBCM. ECM remodelling pathways were enriched in PBMC from COVID-19 patients compared to healthy controls. Patients treated at the intensive care unit (ICU) expressed distinct ECM remodelling gene profiles compared to patients in the hospital ward. Several markers were strongly correlated to immune cell subsets, and the dysregulation in the ICU patients was positively associated with plasma levels of inflammatory cytokines and negatively associated with B-cell activating factors. Finally, our analysis of publicly accessible datasets revealed (i) an augmented ECM remodelling signature in inflamed lung tissue compared to non-inflamed tissue and (ii) proteomics analysis of PBMC from severe COVID-19 patients demonstrated an up-regulation in an ECM remodelling pathway. Our results may suggest the presence of an interaction between ECM remodelling, inflammation, and immune cells, potentially initiating or perpetuating pulmonary pathology in severe COVID-19.


Sujet(s)
COVID-19 , Matrice extracellulaire , Agranulocytes , SARS-CoV-2 , Humains , COVID-19/immunologie , COVID-19/sang , Agranulocytes/immunologie , Agranulocytes/métabolisme , Matrice extracellulaire/métabolisme , Mâle , Femelle , Adulte d'âge moyen , SARS-CoV-2/physiologie , SARS-CoV-2/immunologie , Sujet âgé , Cytokines/sang , Protéomique/méthodes , Poumon/immunologie , Poumon/anatomopathologie , Adulte
5.
Adv Protein Chem Struct Biol ; 141: 361-380, 2024.
Article de Anglais | MEDLINE | ID: mdl-38960480

RÉSUMÉ

Multicellular organisms consist of cells and extracellular matrix (ECM). ECM creates a cellular microenvironment, and cells locally degrade the ECM according to their cellular activity. A major group of enzymes that modify ECM belongs to matrix metalloproteinases (MMPs) and play major roles in various pathophysiological events. ECM degradation by MMPs does not occur in all cellular surroundings but only where it is necessary, and cells achieve this by directionally secreting these proteolytic enzymes. Recent studies have indicated that such enzyme secretion is achieved by targeted vesicle transport along the microtubules, and several kinesin superfamily proteins (KIFs) have been identified as responsible motor proteins involved in the processes. This chapter discusses recent findings of the vesicle transport of MMPs and their roles.


Sujet(s)
Matrix metalloproteinases , Matrix metalloproteinases/métabolisme , Humains , Animaux , Kinésine/métabolisme , Kinésine/composition chimique , Matrice extracellulaire/métabolisme , Transport biologique , Microtubules/métabolisme
6.
Sci Rep ; 14(1): 15022, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38951570

RÉSUMÉ

Cartilage tissue engineering aims to develop functional substitutes for treating cartilage defects and osteoarthritis. Traditional two-dimensional (2D) cell culture systems lack the complexity of native cartilage, leading to the development of 3D regenerative cartilage models. In this study, we developed a 3D model using Gelatin Methacryloyl (GelMA)-based hydrogels seeded with Y201 cells, a bone marrow mesenchymal stem cell line. The model investigated chondrogenic differentiation potential in response to Wnt3a stimulation within the GelMA scaffold and validated using known chondrogenic agonists. Y201 cells demonstrated suitability for the model, with increased proteoglycan content and upregulated chondrogenic marker expression under chondrogenic conditions. Wnt3a enhanced cell proliferation, indicating activation of the Wnt/ß-catenin pathway, which plays a role in cartilage development. GelMA hydrogels provided an optimal scaffold, supporting cell viability and proliferation. The 3D model exhibited consistent responses to chondrogenic agonists, with TGF-ß3 enhancing cartilage-specific extracellular matrix (ECM) production and chondrogenic differentiation. The combination of Wnt3a and TGF-ß3 showed synergistic effects, promoting chondrogenic differentiation and ECM production. This study presents a 3D regenerative cartilage model with potential for investigating cartilage biology, disease mechanisms, and drug screening. The model provides insights into complex cartilage regeneration mechanisms and offers a platform for developing therapeutic approaches for cartilage repair and osteoarthritis treatment.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Chondrogenèse , Hydrogels , Cellules souches mésenchymateuses , Ingénierie tissulaire , Protéine Wnt3A , Protéine Wnt3A/métabolisme , Chondrogenèse/effets des médicaments et des substances chimiques , Ingénierie tissulaire/méthodes , Prolifération cellulaire/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Humains , Cartilage/métabolisme , Gélatine/composition chimique , Structures d'échafaudage tissulaires/composition chimique , Facteur de croissance transformant bêta-3/métabolisme , Facteur de croissance transformant bêta-3/pharmacologie , Lignée cellulaire , Matrice extracellulaire/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/cytologie , Animaux
7.
Sci Rep ; 14(1): 15304, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961188

RÉSUMÉ

Inflammation, corticosteroids, and loading all affect tendon healing, with an interaction between them. However, underlying mechanisms behind the effect of corticosteroids and the interaction with loading remain unclear. The aim of this study was to investigate the role of dexamethasone during tendon healing, including specific effects on tendon cells. Rats (n = 36) were randomized to heavy loading or mild loading, the Achilles tendon was transected, and animals were treated with dexamethasone or saline. Gene and protein analyses of the healing tendon were performed for extracellular matrix-, inflammation-, and tendon cell markers. We further tested specific effects of dexamethasone on tendon cells in vitro. Dexamethasone increased mRNA levels of S100A4 and decreased levels of ACTA2/α-SMA, irrespective of load level. Heavy loading + dexamethasone reduced mRNA levels of FN1 and TenC (p < 0.05), while resolution-related genes were unaltered (p > 0.05). In contrast, mild loading + dexamethasone increased mRNA levels of resolution-related genes ANXA1, MRC1, PDPN, and PTGES (p < 0.03). Altered protein levels were confirmed in tendons with mild loading. Dexamethasone treatment in vitro prevented tendon construct formation, increased mRNA levels of S100A4 and decreased levels of SCX and collagens. Dexamethasone during tendon healing appears to act through immunomodulation by promoting resolution, but also through an effect on tendon cells.


Sujet(s)
Tendon calcanéen , Dexaméthasone , Traumatismes des tendons , Cicatrisation de plaie , Dexaméthasone/pharmacologie , Animaux , Rats , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Traumatismes des tendons/traitement médicamenteux , Traumatismes des tendons/métabolisme , Tendon calcanéen/effets des médicaments et des substances chimiques , Tendon calcanéen/métabolisme , Tendon calcanéen/traumatismes , Tendon calcanéen/anatomopathologie , Protéine S100A4 liant le calcium/métabolisme , Protéine S100A4 liant le calcium/génétique , Mâle , Annexine A1/métabolisme , Annexine A1/génétique , Actines/métabolisme , Actines/génétique , Collagène/métabolisme , Rat Sprague-Dawley , Tendons/effets des médicaments et des substances chimiques , Tendons/métabolisme , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , ARN messager/métabolisme , ARN messager/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice
8.
Proc Natl Acad Sci U S A ; 121(28): e2317711121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38968101

RÉSUMÉ

Adult neural stem cells (NSCs) reside in the dentate gyrus of the hippocampus, and their capacity to generate neurons and glia plays a role in learning and memory. In addition, neurodegenerative diseases are known to be caused by a loss of neurons and glial cells, resulting in a need to better understand stem cell fate commitment processes. We previously showed that NSC fate commitment toward a neuronal or glial lineage is strongly influenced by extracellular matrix stiffness, a property of elastic materials. However, tissues in vivo are not purely elastic and have varying degrees of viscous character. Relatively little is known about how the viscoelastic properties of the substrate impact NSC fate commitment. Here, we introduce a polyacrylamide-based cell culture platform that incorporates mismatched DNA oligonucleotide-based cross-links as well as covalent cross-links. This platform allows for tunable viscous stress relaxation properties via variation in the number of mismatched base pairs. We find that NSCs exhibit increased astrocytic differentiation as the degree of stress relaxation is increased. Furthermore, culturing NSCs on increasingly stress-relaxing substrates impacts cytoskeletal dynamics by decreasing intracellular actin flow rates and stimulating cyclic activation of the mechanosensitive protein RhoA. Additionally, inhibition of motor-clutch model components such as myosin II and focal adhesion kinase partially or completely reverts cells to lineage distributions observed on elastic substrates. Collectively, our results introduce a unique system for controlling matrix stress relaxation properties and offer insight into how NSCs integrate viscoelastic cues to direct fate commitment.


Sujet(s)
Différenciation cellulaire , Cellules souches neurales , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Cellules souches neurales/physiologie , Animaux , Astrocytes/cytologie , Astrocytes/métabolisme , Astrocytes/physiologie , Souris , Résines acryliques/composition chimique , Protéine G RhoA/métabolisme , Cellules cultivées , Neurones/métabolisme , Neurones/physiologie , Neurones/cytologie , Matrice extracellulaire/métabolisme , Contrainte mécanique
9.
Sci Adv ; 10(27): eado9120, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38959311

RÉSUMÉ

A bioinspired hydrogel composed of hyaluronic acid-graft-dopamine (HADA) and a designer peptide HGF-(RADA)4-DGDRGDS (HRR) was presented to enhance tissue integration following spinal cord injury (SCI). The HADA/HRR hydrogel manipulated the infiltration of PDGFRß+ cells in a parallel pattern, transforming dense scars into an aligned fibrous substrate that guided axonal regrowth. Further incorporation of NT3 and curcumin promoted axonal regrowth and survival of interneurons at lesion borders, which served as relays for establishing heterogeneous axon connections in a target-specific manner. Notable improvements in motor, sensory, and bladder functions resulted in rats with complete spinal cord transection. The HADA/HRR + NT3/Cur hydrogel promoted V2a neuron accumulation in ventral spinal cord, facilitating the recovery of locomotor function. Meanwhile, the establishment of heterogeneous neural connections across the hemisected lesion of canines was documented in a target-specific manner via neuronal relays, significantly improving motor functions. Therefore, biomaterials can inspire beneficial biological activities for SCI repair.


Sujet(s)
Matrice extracellulaire , Hydrogels , Traumatismes de la moelle épinière , Traumatismes de la moelle épinière/métabolisme , Traumatismes de la moelle épinière/anatomopathologie , Animaux , Hydrogels/composition chimique , Rats , Matrice extracellulaire/métabolisme , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Chiens , Axones/métabolisme , Axones/effets des médicaments et des substances chimiques , Régénération nerveuse/effets des médicaments et des substances chimiques , Acide hyaluronique/composition chimique , Acide hyaluronique/métabolisme , Récupération fonctionnelle/effets des médicaments et des substances chimiques , Dopamine/métabolisme , Femelle , Modèles animaux de maladie humaine , Rat Sprague-Dawley , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Moelle spinale/métabolisme
10.
Invest Ophthalmol Vis Sci ; 65(8): 1, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38949632

RÉSUMÉ

Purpose: Glucocorticoid-induced glaucoma (GIG) is a prevalent complication associated with glucocorticoids (GCs), resulting in irreversible blindness. GIG is characterized by the abnormal deposition of extracellular matrix (ECM) in the trabecular meshwork (TM), elevation of intraocular pressure (IOP), and loss of retinal ganglion cells (RGCs). The objective of this study is to investigate the effects of nicotinamide riboside (NR) on TM in GIG. Methods: Primary human TM cells (pHTMs) and C57BL/6J mice responsive to GCs were utilized to establish in vitro and in vivo GIG models, respectively. The study assessed the expression of ECM-related proteins in TM and the functions of pHTMs to reflect the effects of NR. Mitochondrial morphology and function were also examined in the GIG cell model. GIG progression was monitored through IOP, RGCs, and mitochondrial morphology. Intracellular nicotinamide adenine dinucleotide (NAD+) levels of pHTMs were enzymatically assayed. Results: NR significantly prevented the expression of ECM-related proteins and alleviated dysfunction in pHTMs after dexamethasone treatment. Importantly, NR protected damaged ATP synthesis, preventing overexpression of mitochondrial reactive oxygen species (ROS), and also protect against decreased mitochondrial membrane potential induced by GCs in vitro. In the GIG mouse model, NR partially prevented the elevation of IOP and the loss of RGCs. Furthermore, NR effectively suppressed the excessive expression of ECM-associated proteins and mitigated mitochondrial damage in vivo. Conclusions: Based on the results, NR effectively enhances intracellular levels of NAD+, thereby mitigating abnormal ECM deposition and TM dysfunction in GIG by attenuating mitochondrial damage induced by GCs. Thus, NR has promising potential as a therapeutic candidate for GIG treatment.


Sujet(s)
Modèles animaux de maladie humaine , Matrice extracellulaire , Glaucome , Glucocorticoïdes , Pression intraoculaire , Souris de lignée C57BL , Mitochondries , Nicotinamide , Composés de pyridinium , Réseau trabéculaire de la sclère , Animaux , Nicotinamide/analogues et dérivés , Nicotinamide/pharmacologie , Composés de pyridinium/pharmacologie , Glucocorticoïdes/toxicité , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Souris , Glaucome/métabolisme , Glaucome/traitement médicamenteux , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Pression intraoculaire/effets des médicaments et des substances chimiques , Humains , Réseau trabéculaire de la sclère/métabolisme , Réseau trabéculaire de la sclère/effets des médicaments et des substances chimiques , Réseau trabéculaire de la sclère/anatomopathologie , Cellules cultivées , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Dexaméthasone/pharmacologie , Mâle
11.
Hepatol Commun ; 8(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967589

RÉSUMÉ

BACKGROUND: Novel noninvasive predictors of disease severity and prognosis in primary sclerosing cholangitis (PSC) are needed. This study evaluated the ability of extracellular matrix remodeling markers to diagnose fibrosis stage and predict PSC-related fibrosis progression and clinical events. METHODS: Liver histology and serum markers of collagen formation (propeptide of type III collagen [Pro-C3], propeptide of type IV collagen, propeptide of type V collagen), collagen degradation (type III collagen matrix metalloproteinase degradation product and type IV collagen matrix metalloproteinase degradation product), and fibrosis (enhanced liver fibrosis [ELF] score and its components [metalloproteinase-1, type III procollagen, hyaluronic acid]) were assessed in samples from baseline to week 96 in patients with PSC enrolled in a study evaluating simtuzumab (NCT01672853). Diagnostic performance for advanced fibrosis (Ishak stages 3-6) and cirrhosis (Ishak stages 5-6) was evaluated by logistic regression and AUROC. Prognostic performance for PSC-related clinical events and fibrosis progression was assessed by AUROC and Wilcoxon rank-sum test. RESULTS: Among 234 patients, 51% had advanced fibrosis and 11% had cirrhosis at baseline. Baseline Pro-C3 and ELF score and its components provided moderate diagnostic ability for discrimination of advanced fibrosis (AUROC 0.73-0.78) and cirrhosis (AUROC 0.73-0.81). Baseline Pro-C3, ELF score, and type III procollagen provided a moderate prognosis for PSC-related clinical events (AUROC 0.70-0.71). Among patients without cirrhosis at baseline, median changes in Pro-C3 and ELF score to week 96 were higher in those with than without progression to cirrhosis (both p < 0.001). CONCLUSIONS: Pro-C3 correlated with fibrosis stage, and Pro-C3 and ELF score provided discrimination of advanced fibrosis and cirrhosis and predicted PSC-related events and fibrosis progression. The results support the clinical utility of Pro-C3 and ELF score for staging and as prognostic markers in PSC.


Sujet(s)
Anticorps monoclonaux humanisés , Marqueurs biologiques , Angiocholite sclérosante , Évolution de la maladie , Matrice extracellulaire , Cirrhose du foie , Humains , Angiocholite sclérosante/traitement médicamenteux , Angiocholite sclérosante/sang , Angiocholite sclérosante/anatomopathologie , Mâle , Femelle , Marqueurs biologiques/sang , Pronostic , Adulte , Cirrhose du foie/sang , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/étiologie , Anticorps monoclonaux humanisés/usage thérapeutique , Adulte d'âge moyen , Matrice extracellulaire/anatomopathologie , Indice de gravité de la maladie , Acide hyaluronique/sang , Foie/anatomopathologie
12.
Sci Rep ; 14(1): 15556, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969656

RÉSUMÉ

Previously, we reported successful cellular expansion of a murine colorectal carcinoma cell line (CT-26) using a three-dimensional (3D) engineered extracellular matrix (EECM) fibrillar scaffold structure. CCL-247 were grown over a limited time period of 8 days on 3D EECM or tissue culture polystyrene (TCPS). Cells were then assayed for growth, electroporation efficiency and Vigil manufacturing release criteria. Using EECM scaffolds, we report an expansion of CCL-247 (HCT116), a colorectal carcinoma cell line, from a starting concentration of 2.45 × 105 cells to 1.9 × 106 cells per scaffold. Following expansion, 3D EECM-derived cells were assessed based on clinical release criteria of the Vigil manufacturing process utilized for Phase IIb trial operation with the FDA. 3D EECM-derived cells passed all Vigil manufacturing release criteria including cytokine expression. Here, we demonstrate successful Vigil product manufacture achieving the specifications necessary for the clinical trial product release of Vigil treatment. Our results confirm that 3D EECM can be utilized for the expansion of human cancer cell CCL-247, justifying further clinical development involving human tissue sample manufacturing including core needle biopsy and minimal ascites samples.


Sujet(s)
Matrice extracellulaire , Immunothérapie , Structures d'échafaudage tissulaires , Humains , Structures d'échafaudage tissulaires/composition chimique , Immunothérapie/méthodes , Ingénierie tissulaire/méthodes , Cellules HCT116 , Tumeurs colorectales/anatomopathologie , Animaux , Souris , Prolifération cellulaire , Lignée cellulaire tumorale , Techniques de cultures cellulaires tridimensionnelles/méthodes
13.
Cell Mol Neurobiol ; 44(1): 54, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969910

RÉSUMÉ

The extracellular matrix (ECM) is a dynamic set of molecules produced by the cellular component of normal and pathological tissues of the embryo and adult. ECM acts as critical regulator in various biological processes such as differentiation, cell proliferation, angiogenesis, and immune control. The most frequent primary brain tumors are gliomas and by far the majority are adult astrocytic tumors (AATs). The prognosis for patients with these neoplasms is poor and the treatments modestly improves survival. In the literature, there is a fair number of studies concerning the composition of the ECM in AATs, while the number of studies relating the composition of the ECM with the immune regulation is smaller. Circulating ECM proteins have emerged as a promising biomarker that reflect the general immune landscape of tumor microenvironment and may represent a useful tool in assessing disease activity. Given the importance it can have for therapeutic and prognostic purposes, the aim of our study is to summarize the biological properties of ECM components and their effects on the tumor microenvironment and to provide an overview of the interactions between major ECM proteins and immune cells in AATs. As the field of immunotherapy in glioma is quickly expanding, we retain that current data together with future studies on ECM organization and functions in glioma will provide important insights into the tuning of immunotherapeutic approaches.


Sujet(s)
Astrocytome , Matrice extracellulaire , Microenvironnement tumoral , Humains , Matrice extracellulaire/métabolisme , Microenvironnement tumoral/immunologie , Astrocytome/anatomopathologie , Astrocytome/métabolisme , Astrocytome/immunologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/métabolisme , Adulte , Animaux , Protéines de la matrice extracellulaire/métabolisme
14.
J Ovarian Res ; 17(1): 139, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970048

RÉSUMÉ

Ovarian fibrosis, characterized by the excessive proliferation of ovarian fibroblasts and the accumulation of extracellular matrix (ECM), serves as one of the primary causes of ovarian dysfunction. Despite the critical role of ovarian fibrosis in maintaining the normal physiological function of the mammalian ovaries, research on this condition has been greatly underestimated, which leads to a lack of clinical treatment options for ovarian dysfunction caused by fibrosis. This review synthesizes recent research on the molecular mechanisms of ovarian fibrosis, encompassing TGF-ß, extracellular matrix, inflammation, and other profibrotic factors contributing to abnormal ovarian fibrosis. Additionally, we summarize current treatment approaches for ovarian dysfunction targeting ovarian fibrosis, including antifibrotic drugs, stem cell transplantation, and exosomal therapies. The purpose of this review is to summarize the research progress on ovarian fibrosis and to propose potential therapeutic strategies targeting ovarian fibrosis for the treatment of ovarian dysfunction.


Sujet(s)
Fibrose , Ovaire , Humains , Femelle , Ovaire/anatomopathologie , Ovaire/métabolisme , Animaux , Matrice extracellulaire/métabolisme , Maladies ovariennes/métabolisme , Maladies ovariennes/anatomopathologie , Maladies ovariennes/thérapie , Thérapie moléculaire ciblée , Facteur de croissance transformant bêta/métabolisme
15.
Cell Death Dis ; 15(6): 455, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937435

RÉSUMÉ

ADGRF5 (GPR116) has been identified as a facilitator of breast cancer cell migration and metastasis, yet the underlying mechanisms remain largely elusive. Our current study reveals that the absence of ADGRF5 in breast cancer cells impairs extracellular matrix (ECM)-associated cell motility and impedes in vivo tumor growth. This correlates with heightened expression of matrix metalloproteinase 8 (MMP8), a well-characterized antitumorigenic MMP, and a shift in the polarization of tumor-associated neutrophils (TANs) towards the antitumor N1 phenotype in the tumor microenvironment (TME). Mechanistically, ADGRF5 inhibits ERK1/2 activity by enhancing RhoA activation, leading to decreased phosphorylation of C/EBPß at Thr235, hindering its nuclear translocation and subsequent activation. Crucially, two C/EBPß binding motifs essential for MMP8 transcription are identified within its promoter region. Consequently, ADGRF5 silencing fosters MMP8 expression and CXCL8 secretion, attracting increased infiltration of TANs; simultaneously, MMP8 plays a role in decorin cleavage, which leads to trapped-inactivation of TGF-ß in the TME, thereby polarizing TANs towards the antitumor N1 neutrophil phenotype and mitigating TGF-ß-enhanced cell motility in breast cancer. Our findings reveal a novel connection between ADGRF5, an adhesion G protein-coupled receptor, and the orchestration of the TME, which dictates malignancy progression. Overall, the data underscore ADGRF5 as a promising therapeutic target for breast cancer intervention.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Matrix metalloproteinase 8 , Récepteurs couplés aux protéines G , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Matrix metalloproteinase 8/métabolisme , Matrix metalloproteinase 8/génétique , Animaux , Récepteurs couplés aux protéines G/métabolisme , Lignée cellulaire tumorale , Microenvironnement tumoral , Souris , Évolution de la maladie , Granulocytes neutrophiles/métabolisme , Souris nude , Protéine G RhoA/métabolisme , Interleukine-8/métabolisme , Régulation de l'expression des gènes tumoraux , Facteur de croissance transformant bêta/métabolisme , Matrice extracellulaire/métabolisme
16.
J Extracell Vesicles ; 13(7): e12461, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38940266

RÉSUMÉ

Cardiac fibrosis is a common pathological feature of cardiovascular diseases that arises from the hyperactivation of fibroblasts and excessive extracellular matrix (ECM) deposition, leading to impaired cardiac function and potentially heart failure or arrhythmia. Extracellular vesicles (EVs) released by cardiomyocytes (CMs) regulate various physiological functions essential for myocardial homeostasis, which are disrupted in cardiac disease. Therefore, healthy CM-derived EVs represent a promising cell-free therapy for the treatment of cardiac fibrosis. To this end, we optimized the culture conditions of human adult CMs to obtain a large yield of EVs without compromising cellular integrity by using a defined combination of small molecules. EVs were isolated by ultracentrifugation, and their characteristics were analysed. Finally, their effect on fibrosis was tested. Treatment of TGFß-activated human cardiac fibroblasts with EVs derived from CMs using our culture system resulted in a decrease in fibroblast activation markers and ECM accumulation. The rescued phenotype was associated with specific EV cargo, including multiple myocyte-specific and antifibrotic microRNAs, although their effect individually was not as effective as the EV treatment. Notably, pathway analysis showed that EV treatment reverted the transcription of activated fibroblasts and decreased several signalling pathways, including MAPK, mTOR, JAK/STAT, TGFß, and PI3K/Akt, all of which are involved in fibrosis development. Intracardiac injection of CM-derived EVs in an animal model of cardiac fibrosis reduced fibrotic area and increased angiogenesis, which correlated with improved cardiac function. These findings suggest that EVs derived from human adult CMs may offer a targeted and effective treatment for cardiac fibrosis, owing to their antifibrotic properties and the specificity of cargo.


Sujet(s)
Vésicules extracellulaires , Fibrose , Myocytes cardiaques , Myocytes cardiaques/métabolisme , Humains , Vésicules extracellulaires/métabolisme , Fibroblastes/métabolisme , Animaux , microARN/métabolisme , Matrice extracellulaire/métabolisme , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Cellules cultivées , Souris , Adulte
17.
J Nanobiotechnology ; 22(1): 378, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38943185

RÉSUMÉ

Tissue engineered heart valves (TEHVs) demonstrates the potential for tissue growth and remodel, offering particular benefit for pediatric patients. A significant challenge in designing functional TEHV lies in replicating the anisotropic mechanical properties of native valve leaflets. To establish a biomimetic TEHV model, we employed melt-electrowriting (MEW) technology to fabricate an anisotropic PCL scaffold. By integrating the anisotropic MEW-PCL scaffold with bioactive hydrogels (GelMA/ChsMA), we successfully crafted an elastic scaffold with tunable mechanical properties closely mirroring the structure and mechanical characteristics of natural heart valves. This scaffold not only supports the growth of valvular interstitial cells (VICs) within a 3D culture but also fosters the remodeling of extracellular matrix of VICs. The in vitro experiments demonstrated that the introduction of ChsMA improved the hemocompatibility and endothelialization of TEHV scaffold. The in vivo experiments revealed that, compared to their non-hydrogel counterparts, the PCL-GelMA/ChsMA scaffold, when implanted into SD rats, significantly suppressed immune reactions and calcification. In comparison with the PCL scaffold, the PCL-GelMA/ChsMA scaffold exhibited higher bioactivity and superior biocompatibility. The amalgamation of MEW technology and biomimetic design approaches provides a new paradigm for manufacturing scaffolds with highly controllable microstructures, biocompatibility, and anisotropic mechanical properties required for the fabrication of TEHVs.


Sujet(s)
Valves cardiaques , Hydrogels , Rat Sprague-Dawley , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Ingénierie tissulaire/méthodes , Animaux , Structures d'échafaudage tissulaires/composition chimique , Anisotropie , Rats , Hydrogels/composition chimique , Matériaux biocompatibles/composition chimique , Prothèse valvulaire cardiaque , Polyesters/composition chimique , Cellules cultivées , Humains , Matrice extracellulaire/composition chimique , Mâle
18.
J Orthop Surg Res ; 19(1): 377, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38926735

RÉSUMÉ

BACKGROUND: Achilles tendon is vital in maintaining the stability and function of ankle joint. It is quite difficult to achieve the structural and functional repair of Achilles tendon in tissue engineering. METHODS: A tissue-engineered tendon micro-tissue was prepared using rat tail tendon extracellular matrix (TECM) combined with rat adipose stem cells (ADSCs) to repair Achilles tendon injuries. The TECM was prepared by repeated freezing and thawing. The in vitro characteristics of TECM and its effect on ADSCs proliferation were detected. This tissue-engineered tendon micro-tissue for Achilles tendon repair in vivo was evaluated based on general characteristics, gait analysis, ultrasound findings, histological analysis, and biomechanical testing. RESULTS: The results showed that the TECM scaffold had good biocompatibility for ADSCs. At 2 weeks post-surgery, collagen types I and III and tenomodulin expression were higher, and vascular endothelial growth factor expression was lower in the micro-tissue group than other groups. At 4 and 8 weeks post-surgery, the results of histological analysis and ultrasound findings showed that the repaired tendon tissue was smooth and lustrous, and was arranged regularly and evenly in the micro-tissue group. Gait analysis confirmed that better motor function recovery was noted in micro-tissue group than other groups. In addition, the mechanical properties of the repaired tendon tissue in micro-tissue group were better than other groups. CONCLUSION: Tissue-engineered tendon micro-tissue fabricated by TECM and ADSCs has good biocompatibility and can promote structural and functional repair of tendon in vivo. This composite biomaterial has broad application prospects in tissue engineering.


Sujet(s)
Tendon calcanéen , Matrice extracellulaire , Rat Sprague-Dawley , Régénération , Traumatismes des tendons , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Animaux , Ingénierie tissulaire/méthodes , Tendon calcanéen/traumatismes , Tendon calcanéen/physiologie , Traumatismes des tendons/thérapie , Régénération/physiologie , Rats , Mâle , Tissu adipeux/cytologie
19.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38928032

RÉSUMÉ

Bowel strictures are well recognized as one of the most severe complications in Crohn's disease, with variable impacts on the prognosis and often needing surgical or endoscopic treatment. Distinguishing inflammatory strictures from fibrotic ones is of primary importance due to the different therapeutic approaches required. Indeed, to better understand the pathogenesis of fibrosis, it is crucial to investigate molecular processes involving genetic factors, cytokines, alteration of the intestinal barrier, and epithelial and endothelial damage, leading to an increase in extracellular matrix synthesis, which ultimately ends in fibrosis. In such a complex mechanism, the gut microbiota also seems to play a role. A better comprehension of molecular processes underlying bowel fibrosis, in addition to radiological and histopathological findings, has led to the identification of high-risk patients for personalized follow-up and testing of new therapies, primarily in preclinical models, targeting specific pathways involving Transforming Growth Factor-ß, interleukins, extracellular matrix balance, and gut microbiota. Our review aims to summarize current evidence about molecular factors involved in intestinal fibrosis' pathogenesis, paving the way for potential diagnostic biomarkers or anti-fibrotic treatments for stricturing Crohn's disease.


Sujet(s)
Maladie de Crohn , Fibrose , Microbiome gastro-intestinal , Humains , Maladie de Crohn/métabolisme , Maladie de Crohn/anatomopathologie , Maladie de Crohn/thérapie , Animaux , Matrice extracellulaire/métabolisme , Marqueurs biologiques , Cytokines/métabolisme
20.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38928148

RÉSUMÉ

Investigate meniscal extracellular matrix degradation. Equine menisci (n = 34 from 17 horses) were studied. Site-matched sections were cut and scored from three regions (ROIs; n = 102) and stained for histology, proteoglycan (safranin O and fast green), aggrecan, and collagen cleavage (NITEGE, DIPEN, and C1,2C antibodies, respectively). Picrosirius red and second harmonic generation microscopy were performed to investigate collagen ultrastructure. A total of 42 ROIs met the inclusion criteria and were included in the final analysis. The median (range) ROI histological score was 3 (0-9), providing a large spectrum of pathology. The median (range) proteoglycan score was 1 (0-3), representing superficial and central meniscal loss. The median (range) of DIPEN, NITEGE, and C1,2C scores was 1 (0-3), revealing immunostaining of the femoral and tibial surfaces. The proteoglycan scores exhibited significant positive associations with both histologic evaluation (p = 0.03) and DIPEN scores (p = 0.02). Additionally, a robust positive association (p = 0.007) was observed between the two aggrecanolysis indicators, NITEGE and DIPEN scores. A negative association (p = 0.008) was identified between NITEGE and histological scores. The C1,2C scores were not associated with any other scores. Picrosirius red and second harmonic generation microscopy (SHGM) illustrated the loss of the collagen matrix and structure centrally. Proteoglycan and collagen degradation commonly occur superficially in menisci and less frequently centrally. The identification of central meniscal proteoglycan and collagen degradation provides novel insight into central meniscal degeneration. However, further research is needed to elucidate the etiology and sequence of degradative events.


Sujet(s)
Collagène , Ménisque , Protéoglycanes , Animaux , Equus caballus , Protéoglycanes/métabolisme , Collagène/métabolisme , Ménisque/métabolisme , Agrécanes/métabolisme , Matrice extracellulaire/métabolisme , Protéolyse , Ménisques de l'articulation du genou/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...