Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 14.031
Filtrer
1.
Front Immunol ; 15: 1385006, 2024.
Article de Anglais | MEDLINE | ID: mdl-38895122

RÉSUMÉ

Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.


Sujet(s)
Fibroblastes , Fibrose , Arthrose , Phénotype , Cellules synoviales , Humains , Arthrose/anatomopathologie , Arthrose/immunologie , Arthrose/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Animaux , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Cellules synoviales/immunologie , Membrane synoviale/anatomopathologie , Membrane synoviale/immunologie , Membrane synoviale/métabolisme
2.
PeerJ ; 12: e17551, 2024.
Article de Anglais | MEDLINE | ID: mdl-38887622

RÉSUMÉ

Background: Keloid is a chronic proliferative fibrotic disease caused by abnormal fibroblasts proliferation and excessive extracellular matrix (ECM) production. Numerous fibrotic disorders are significantly influenced by ferroptosis, and targeting ferroptosis can effectively mitigate fibrosis development. This study aimed to investigate the role and mechanism of ferroptosis in keloid development. Methods: Keloid tissues from keloid patients and normal skin tissues from healthy controls were collected. Iron content, lipid peroxidation (LPO) level, and the mRNA and protein expression of ferroptosis-related genes including solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), transferrin receptor (TFRC), and nuclear factor erythroid 2-related factor 2 (Nrf2) were determined. Mitochondrial morphology was observed using transmission electron microscopy (TEM). Keloid fibroblasts (KFs) were isolated from keloid tissues, and treated with ferroptosis inhibitor ferrostatin-1 (fer-1) or ferroptosis activator erastin. Iron content, ferroptosis-related marker levels, LPO level, mitochondrial membrane potential, ATP content, and mitochondrial morphology in KFs were detected. Furthermore, the protein levels of α-smooth muscle actin (α-SMA), collagen I, and collagen III were measured to investigate whether ferroptosis affect fibrosis in KFs. Results: We found that iron content and LPO level were substantially elevated in keloid tissues and KFs. SLC7A11, GPX4, and Nrf2 were downregulated and TFRC was upregulated in keloid tissues and KFs. Mitochondria in keloid tissues and KFs exhibited ferroptosis-related pathology. Fer-1 treatment reduced iron content, restrained ferroptosis and mitochondrial dysfunction in KFs, Moreover, ferrostatin-1 restrained the protein expression of α-SMA, collagen I, and collagen III in KFs. Whereas erastin treatment showed the opposite results. Conclusion: Ferroptosis exists in keloid. Ferrostatin-1 restrained ECM deposition and fibrosis in keloid through inhibiting ferroptosis, and erastin induced ECM deposition and fibrosis through intensifying ferroptosis.


Sujet(s)
Cyclohexylamines , Ferroptose , Fibroblastes , Fibrose , Chéloïde , Facteur-2 apparenté à NF-E2 , Phénylènediamines , Phospholipid hydroperoxide glutathione peroxidase , Humains , Ferroptose/effets des médicaments et des substances chimiques , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Chéloïde/traitement médicamenteux , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Cyclohexylamines/pharmacologie , Fibrose/métabolisme , Fibrose/anatomopathologie , Phénylènediamines/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Mâle , Peroxydation lipidique/effets des médicaments et des substances chimiques , Femelle , Adulte , Fer/métabolisme , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Pipérazines/pharmacologie , Actines/métabolisme , Actines/génétique , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques
3.
Cells ; 13(11)2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38891046

RÉSUMÉ

Fibroblasts, among the most prevalent and widely distributed cell types in the human body, play a crucial role in defining tissue structure. They do this by depositing and remodeling extracellular matrixes and organizing functional tissue networks, which are essential for tissue homeostasis and various human diseases. Pulmonary hypertension (PH) is a devastating syndrome with high mortality, characterized by remodeling of the pulmonary vasculature and significant cellular and structural changes within the intima, media, and adventitia layers. Most research on PH has focused on alterations in the intima (endothelial cells) and media (smooth muscle cells). However, research over the past decade has provided strong evidence of the critical role played by pulmonary artery adventitial fibroblasts in PH. These fibroblasts exhibit the earliest, most dramatic, and most sustained proliferative, apoptosis-resistant, and inflammatory responses to vascular stress. This review examines the aberrant phenotypes of PH fibroblasts and their role in the pathogenesis of PH, discusses potential molecular signaling pathways underlying these activated phenotypes, and highlights areas of research that merit further study to identify promising targets for the prevention and treatment of PH.


Sujet(s)
Fibroblastes , Hypertension pulmonaire , Humains , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/physiopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Animaux , Transduction du signal , Artère pulmonaire/anatomopathologie , Artère pulmonaire/métabolisme
4.
J Transl Med ; 22(1): 560, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867219

RÉSUMÉ

BACKGROUND: Cardiac fibrosis after myocardial infarction (MI) has been considered an important part of cardiac pathological remodeling. Immune cells, especially macrophages, are thought to be involved in the process of fibrosis and constitute a niche with fibroblasts to promote fibrosis. However, the diversity and variability of fibroblasts and macrophages make it difficult to accurately depict interconnections. METHODS: We collected and reanalyzed scRNA-seq and snRNA-seq datasets from 12 different studies. Differentiation trajectories of these subpopulations after MI injury were analyzed by using scVelo, PAGA and Slingshot. We used CellphoneDB and NicheNet to infer fibroblast-macrophage interactions. Tissue immunofluorescence staining and in vitro experiments were used to validate our findings. RESULTS: We discovered two subsets of ECM-producing fibroblasts, reparative cardiac fibroblasts (RCFs) and matrifibrocytes, which appeared at different times after MI and exhibited different transcriptional profiles. We also observed that CTHRC1+ fibroblasts represent an activated fibroblast in chronic disease states. We identified a macrophage subset expressing the genes signature of SAMs conserved in both human and mouse hearts. Meanwhile, the SPP1hi macrophages were predominantly found in the early stages after MI, and cell communication analysis indicated that SPP1hi macrophage-RCFs interactions are mainly involved in collagen deposition and scar formation. CONCLUSIONS: Overall, this study comprehensively analyzed the dynamics of fibroblast and macrophage subsets after MI and identified specific subsets of fibroblasts and macrophages involved in scar formation and collagen deposition.


Sujet(s)
Fibroblastes , Macrophages , Infarctus du myocarde , Analyse sur cellule unique , Transcriptome , Infarctus du myocarde/génétique , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Macrophages/métabolisme , Animaux , Transcriptome/génétique , Humains , Communication cellulaire , Souris , Différenciation cellulaire/génétique , Souris de lignée C57BL , Myocarde/anatomopathologie , Myocarde/métabolisme , Matrice extracellulaire/métabolisme , Analyse de profil d'expression de gènes
5.
Circ Res ; 134(12): 1703-1717, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843287

RÉSUMÉ

Fibroblasts are essential for building and maintaining the structural integrity of all organs. Moreover, fibroblasts can acquire an inflammatory phenotype to accommodate immune cells in specific niches and to provide migration, differentiation, and growth factors. In the heart, balancing of fibroblast activity is critical for cardiac homeostasis and optimal organ function during inflammation. Fibroblasts sustain cardiac homeostasis by generating local niche environments that support housekeeping functions and by actively engaging in intercellular cross talk. During inflammatory perturbations, cardiac fibroblasts rapidly switch to an inflammatory state and actively communicate with infiltrating immune cells to orchestrate immune cell migration and activity. Here, we summarize the current knowledge on the molecular landscape of cardiac fibroblasts, focusing on their dual role in promoting tissue homeostasis and modulating immune cell-cardiomyocyte interaction. In addition, we discuss potential future avenues for manipulating cardiac fibroblast activity during myocardial inflammation.


Sujet(s)
Fibroblastes , Homéostasie , Myocarde , Humains , Animaux , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Myocarde/anatomopathologie , Myocarde/immunologie , Myocarde/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/immunologie , Myocardite/immunologie , Myocardite/anatomopathologie , Myocardite/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Communication cellulaire
6.
Iran J Allergy Asthma Immunol ; 23(2): 197-220, 2024 Apr 07.
Article de Anglais | MEDLINE | ID: mdl-38822514

RÉSUMÉ

Systemic sclerosis (SSc) is an autoimmune systemic disease that is characterized by immune dysregulation, inflammation, vasculopathy, and fibrosis. Tissue fibrosis plays an important role in SSc and can affect several organs such as the dermis, lungs, and heart. Dysregulation of interferon (IFN) signaling contributes to the SSc pathogenesis and interferon regulatory factor 1 (IRF1) has been indicated as the main regulator of type I IFN. This study aimed to clarify the effect of IFN-gamma (-γ) and dexamethasone (DEX) on the IRF1, extracellular signal-regulated kinase 1/2 (ERK1/2), and the expression of alpha-smooth muscle actin (α-SMA) in myofibroblasts and genes involved in the inflammation and fibrosis processes in early diffuse cutaneous systemic sclerosis (dcSSc). A total of 10 early dcSSc patients (diffuse cutaneous form) and 10 unaffected control dermis biopsies were obtained to determine IFNγ and DEX effects on inflammation and fibrosis. Fibroblasts were treated with IFNγ and DEX at optimum time and dose. The expression level of genes and proteins involved in the fibrosis and inflammation processes have been quantified by quantitative real-time PCR (RT-qPCR) and western blot, respectively. IFNγ could up-regulate some of the inflammation-related genes (Interleukin-6; IL6) and down-regulate some of the fibrosis-related genes (COL1A1) in cultured fibroblasts of patients with early dcSSc compared to the untreated group. Besides, it has been revealed that IFNγ can induce fibroblast differentiation to the myofibroblast that expresses α-SMA. Concerning the inhibitory effect of IFNγ on some fibrotic genes and its positive effect on the inflammatory genes and myofibroblast differentiation, it seems that IFNγ may play a dual role in SSc.


Sujet(s)
Actines , Fibroblastes , Interféron gamma , Interleukine-6 , Sclérodermie systémique , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Actines/métabolisme , Actines/génétique , Cellules cultivées , Dexaméthasone/pharmacologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/effets des médicaments et des substances chimiques , Fibrose , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Facteur-1 de régulation d'interféron/métabolisme , Facteur-1 de régulation d'interféron/génétique , Interféron gamma/pharmacologie , Interleukine-6/métabolisme , Interleukine-6/génétique , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/immunologie
7.
Int J Mol Sci ; 25(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892192

RÉSUMÉ

Cardiac fibrosis, a process characterized by excessive extracellular matrix (ECM) deposition, is a common pathological consequence of many cardiovascular diseases (CVDs) normally resulting in organ failure and death. Cardiac fibroblasts (CFs) play an essential role in deleterious cardiac remodeling and dysfunction. In response to injury, quiescent CFs become activated and adopt a collagen-secreting phenotype highly contributing to cardiac fibrosis. In recent years, studies have been focused on the exploration of molecular and cellular mechanisms implicated in the activation process of CFs, which allow the development of novel therapeutic approaches for the treatment of cardiac fibrosis. Transcriptomic analyses using single-cell RNA sequencing (RNA-seq) have helped to elucidate the high cellular diversity and complex intercellular communication networks that CFs establish in the mammalian heart. Furthermore, a significant body of work supports the critical role of epigenetic regulation on the expression of genes involved in the pathogenesis of cardiac fibrosis. The study of epigenetic mechanisms, including DNA methylation, histone modification, and chromatin remodeling, has provided more insights into CF activation and fibrotic processes. Targeting epigenetic regulators, especially DNA methyltransferases (DNMT), histone acetylases (HAT), or histone deacetylases (HDAC), has emerged as a promising approach for the development of novel anti-fibrotic therapies. This review focuses on recent transcriptomic advances regarding CF diversity and molecular and epigenetic mechanisms that modulate the activation process of CFs and their possible clinical applications for the treatment of cardiac fibrosis.


Sujet(s)
Épigenèse génétique , Fibroblastes , Fibrose , Humains , Animaux , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Méthylation de l'ADN
8.
Cell Mol Life Sci ; 81(1): 264, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38878214

RÉSUMÉ

Atrial fibrillation (AF) is the most common arrhythmia, and atrial fibrosis is a pathological hallmark of structural remodeling in AF. Prostaglandin I2 (PGI2) can prevent the process of fibrosis in various tissues via cell surface Prostaglandin I2 receptor (IP). However, the role of PGI2 in AF and atrial fibrosis remains unclear. The present study aimed to clarify the role of PGI2 in angiotensin II (Ang II)-induced AF and the underlying molecular mechanism. PGI2 content was decreased in both plasma and atrial tissue from patients with AF and mice treated with Ang II. Treatment with the PGI2 analog, iloprost, reduced Ang II-induced AF and atrial fibrosis. Iloprost prevented Ang II-induced atrial fibroblast collagen synthesis and differentiation. RNA-sequencing analysis revealed that iloprost significantly attenuated transcriptome changes in Ang II-treated atrial fibroblasts, especially mitogen-activated protein kinase (MAPK)-regulated genes. We demonstrated that iloprost elevated cAMP levels and then activated protein kinase A, resulting in a suppression of extracellular signal-regulated kinase1/2 and P38 activation, and ultimately inhibiting MAPK-dependent interleukin-6 transcription. In contrast, cardiac fibroblast-specific IP-knockdown mice had increased Ang II-induced AF inducibility and aggravated atrial fibrosis. Together, our study suggests that PGI2/IP system protects against atrial fibrosis and that PGI2 is a therapeutic target for treating AF.The prospectively registered trial was approved by the Chinese Clinical Trial Registry. The trial registration number is ChiCTR2200056733. Data of registration was 2022/02/12.


Sujet(s)
Angiotensine-II , Fibrillation auriculaire , Remodelage auriculaire , Prostacycline , Souris de lignée C57BL , Transduction du signal , Animaux , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/induit chimiquement , Fibrillation auriculaire/prévention et contrôle , Souris , Humains , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Remodelage auriculaire/effets des médicaments et des substances chimiques , Prostacycline/métabolisme , Fibrose , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Atrium du coeur/métabolisme , Atrium du coeur/anatomopathologie , Atrium du coeur/effets des médicaments et des substances chimiques , Iloprost/pharmacologie , Récepteurs de l'époprosténol/métabolisme , Récepteurs de l'époprosténol/génétique , Femelle
9.
Cells ; 13(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38891078

RÉSUMÉ

Pulmonary fibrosis is a chronic, progressive, irreversible lung disease characterized by fibrotic scarring in the lung parenchyma. This condition involves the excessive accumulation of extracellular matrix (ECM) due to the aberrant activation of myofibroblasts in the alveolar environment. Transforming growth factor beta (TGF-ß) signaling is a crucial driver of fibrogenesis because it promotes excessive ECM deposition, thereby leading to scar formation and lung damage. A primary target of TGF-ß signaling in fibrosis is Collagen Triple Helix Repeat Containing 1 (CTHRC1), a secreted glycoprotein that plays a pivotal role in ECM deposition and wound repair. TGF-ß transcriptionally regulates CTHRC1 in response to tissue injury and controls the wound healing response through functional activity. CTHRC1 may also play an essential role in re-establishing and maintaining tissue homeostasis after wound closure by modulating both the TGF-ß and canonical Wnt signaling pathways. This dual function suggests that CTHRC1 regulates tissue remodeling and homeostasis. However, deregulated CTHRC1 expression in pathogenic fibroblasts has recently emerged as a hallmark of fibrosis in multiple organs and tissues. This review highlights recent studies suggesting that CTHRC1 can serve as a diagnostic and prognostic biomarker for fibrosis in idiopathic pulmonary fibrosis, systemic sclerosis, and post-COVID-19 lung fibrosis. Notably, CTHRC1 expression is responsive to antifibrotic drugs that target the TGF-ß pathway, such as pirfenidone and bexotegrast, indicating its potential as a biomarker of treatment success. These findings suggest that CTHRC1 may present new opportunities for diagnosing and treating patients with lung fibrosis.


Sujet(s)
Protéines de la matrice extracellulaire , Fibroblastes , Fibrose pulmonaire , Humains , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Protéines de la matrice extracellulaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Animaux , Facteur de croissance transformant bêta/métabolisme , Matrice extracellulaire/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme
10.
Cells ; 13(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38891103

RÉSUMÉ

Patients with chronic hypoxia show a higher tumor incidence; however, no primary common cause has been recognized. Given the similarities between cellular reprogramming and oncogenic transformation, we directly compared these processes in human cells subjected to hypoxia. Mouse embryonic fibroblasts were employed as controls to compare transfection and reprogramming efficiency; human adipose-derived mesenchymal stem cells were employed as controls in human cells. Easily obtainable human peripheral blood mononuclear cells (PBMCs) were chosen to establish a standard protocol to compare cell reprogramming (into induced pluripotent stem cells (iPSCs)) and oncogenic focus formation efficiency. Cell reprogramming was achieved for all three cell types, generating actual pluripotent cells capable for differentiating into the three germ layers. The efficiencies of the cell reprogramming and oncogenic transformation were similar. Hypoxia slightly increased the reprogramming efficiency in all the cell types but with no statistical significance for PBMCs. Various PBMC types can respond to hypoxia differently; lymphocytes and monocytes were, therefore, reprogrammed separately, finding a significant difference between normoxia and hypoxia in monocytes in vitro. These differences were then searched for in vivo. The iPSCs and oncogenic foci were generated from healthy volunteers and patients with chronic obstructive pulmonary disease (COPD). Although higher iPSC generation efficiency in the patients with COPD was found for lymphocytes, this increase was not statistically significant for oncogenic foci. Remarkably, a higher statistically significant efficiency in COPD monocytes was demonstrated for both processes, suggesting that physiological hypoxia exerts an effect on cell reprogramming and oncogenic transformation in vivo in at least some cell types.


Sujet(s)
Transformation cellulaire néoplasique , Reprogrammation cellulaire , Cellules souches pluripotentes induites , Humains , Reprogrammation cellulaire/génétique , Cellules souches pluripotentes induites/métabolisme , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Animaux , Souris , Hypoxie cellulaire , Agranulocytes/métabolisme , Agranulocytes/cytologie , Mâle , Femelle , Adulte d'âge moyen , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Différenciation cellulaire/génétique , Sujet âgé
11.
Cells ; 13(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38891121

RÉSUMÉ

Hypertension induces cardiac fibrotic remodelling characterised by the phenotypic switching of cardiac fibroblasts (CFs) and collagen deposition. We tested the hypothesis that Wnt1-inducible signalling pathway protein-1 (WISP-1) promotes CFs' phenotypic switch, type I collagen synthesis, and in vivo fibrotic remodelling. The treatment of human CFs (HCFs, n = 16) with WISP-1 (500 ng/mL) induced a phenotypic switch (α-smooth muscle actin-positive) and type I procollagen cleavage to an intermediate form of collagen (pC-collagen) in conditioned media after 24h, facilitating collagen maturation. WISP-1-induced collagen processing was mediated by Akt phosphorylation via integrin ß1, and disintegrin and metalloproteinase with thrombospondin motifs 2 (ADAMTS-2). WISP-1 wild-type (WISP-1+/+) mice and WISP-1 knockout (WISP-1-/-) mice (n = 5-7) were subcutaneously infused with angiotensin II (AngII, 1000 ng/kg/min) for 28 days. Immunohistochemistry revealed the deletion of WISP-1 attenuated type I collagen deposition in the coronary artery perivascular area compared to WISP-1+/+ mice after a 28-day AngII infusion, and therefore, the deletion of WISP-1 attenuated AngII-induced cardiac fibrosis in vivo. Collectively, our findings demonstrated WISP-1 is a critical mediator in cardiac fibrotic remodelling, by promoting CFs' activation via the integrin ß1-Akt signalling pathway, and induced collagen processing and maturation via ADAMTS-2. Thereby, the modulation of WISP-1 levels could provide potential therapeutic targets in clinical treatment.


Sujet(s)
Protéines CCN de signalisation intercellulaire , Fibroblastes , Fibrose , Myocarde , Protéines proto-oncogènes , Animaux , Protéines CCN de signalisation intercellulaire/métabolisme , Protéines CCN de signalisation intercellulaire/génétique , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/effets des médicaments et des substances chimiques , Humains , Souris , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Myocarde/anatomopathologie , Myocarde/métabolisme , Collagène/métabolisme , Angiotensine-II/pharmacologie , Souris knockout , Collagène de type I/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Souris de lignée C57BL
12.
Acta Neuropathol Commun ; 12(1): 90, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851733

RÉSUMÉ

Mitochondrial dysfunctions are key features of Alzheimer's disease (AD). The occurrence of these disturbances in the peripheral cells of AD patients and their potential correlation with disease progression are underinvestigated. We studied mitochondrial structure, function and mitophagy in fibroblasts from healthy volunteers and AD patients at the prodromal (AD-MCI) or demented (AD-D) stages. We carried out correlation studies with clinical cognitive scores, namely, (i) Mini-Mental State Examination (MMSE) and (ii) Dementia Rating-Scale Sum of Boxes (CDR-SOB), and with (iii) amyloid beta (Aß) plaque burden (PiB-PET imaging) and (iv) the accumulation of peripheral amyloid precursor protein C-terminal fragments (APP-CTFs). We revealed alterations in mitochondrial structure as well as specific mitochondrial dysfunction signatures in AD-MCI and AD-D fibroblasts and revealed that defective mitophagy and autophagy are linked to impaired lysosomal activity in AD-D fibroblasts. We reported significant correlations of a subset of these dysfunctions with cognitive decline, AD-related clinical hallmarks and peripheral APP-CTFs accumulation. This study emphasizes the potential use of peripheral cells for investigating AD pathophysiology.


Sujet(s)
Maladie d'Alzheimer , Fibroblastes , Mitochondries , Mitophagie , Humains , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/imagerie diagnostique , Fibroblastes/anatomopathologie , Fibroblastes/métabolisme , Sujet âgé , Femelle , Mitochondries/anatomopathologie , Mitochondries/métabolisme , Mâle , Mitophagie/physiologie , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Précurseur de la protéine bêta-amyloïde/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Dysfonctionnement cognitif/anatomopathologie , Dysfonctionnement cognitif/métabolisme , Autophagie/physiologie
13.
Nature ; 630(8016): 475-483, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839958

RÉSUMÉ

Senescence is a cellular state linked to ageing and age-onset disease across many mammalian species1,2. Acutely, senescent cells promote wound healing3,4 and prevent tumour formation5; but they are also pro-inflammatory, thus chronically exacerbate tissue decline. Whereas senescent cells are active targets for anti-ageing therapy6-11, why these cells form in vivo, how they affect tissue ageing and the effect of their elimination remain unclear12,13. Here we identify naturally occurring senescent glia in ageing Drosophila brains and decipher their origin and influence. Using Activator protein 1 (AP1) activity to screen for senescence14,15, we determine that senescent glia can appear in response to neuronal mitochondrial dysfunction. In turn, senescent glia promote lipid accumulation in non-senescent glia; similar effects are seen in senescent human fibroblasts in culture. Targeting AP1 activity in senescent glia mitigates senescence biomarkers, extends fly lifespan and health span, and prevents lipid accumulation. However, these benefits come at the cost of increased oxidative damage in the brain, and neuronal mitochondrial function remains poor. Altogether, our results map the trajectory of naturally occurring senescent glia in vivo and indicate that these cells link key ageing phenomena: mitochondrial dysfunction and lipid accumulation.


Sujet(s)
Vieillissement , Encéphale , Vieillissement de la cellule , Drosophila melanogaster , Métabolisme lipidique , Mitochondries , Névroglie , Neurones , Animaux , Mitochondries/métabolisme , Névroglie/métabolisme , Névroglie/anatomopathologie , Humains , Drosophila melanogaster/métabolisme , Drosophila melanogaster/cytologie , Encéphale/métabolisme , Encéphale/anatomopathologie , Encéphale/cytologie , Neurones/métabolisme , Neurones/anatomopathologie , Vieillissement/métabolisme , Longévité , Mâle , Femelle , Facteur de transcription AP-1/métabolisme , Stress oxydatif , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie
14.
PLoS One ; 19(6): e0304530, 2024.
Article de Anglais | MEDLINE | ID: mdl-38829908

RÉSUMÉ

Rheumatoid arthritis (RA) is a systemic immune-mediated disease characterized by joint inflammation and destruction. The disease typically affects small joints in the hands and feet, later progressing to involve larger joints such as the knees, shoulders, and hips. While the reasons for these joint-specific differences are unclear, distinct epigenetic patterns associated with joint location have been reported. In this study, we evaluated the unique epigenetic landscapes of fibroblast-like synoviocytes (FLS) from hip and knee synovium in RA patients, focusing on the expression and regulation of Homeobox (HOX) transcription factors. These highly conserved genes play a critical role in embryonic development and are known to maintain distinct expression patterns in various adult tissues. We found that several HOX genes, especially HOXD10, were differentially expressed in knee FLS compared with hip FLS. Epigenetic differences in chromatin accessibility and histone marks were observed in HOXD10 promoter between knee and hip FLS. Histone modification, particularly histone acetylation, was identified as an important regulator of HOXD10 expression. To understand the mechanism of differential HOXD10 expression, we inhibited histone deacetylases (HDACs) with small molecules and siRNA. We found that HDAC1 blockade or deficiency normalized the joint-specific HOXD10 expression patterns. These observations suggest that epigenetic differences, specifically histone acetylation related to increased HDAC1 expression, play a crucial role in joint-specific HOXD10 expression. Understanding these mechanisms could provide insights into the regional aspects of RA and potentially lead to therapeutic strategies targeting specific patterns of joint involvement during the course of disease.


Sujet(s)
Polyarthrite rhumatoïde , Épigenèse génétique , Fibroblastes , Protéines à homéodomaine , Cellules synoviales , Humains , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Régions promotrices (génétique) , Articulation du genou/anatomopathologie , Articulation du genou/métabolisme , Régulation de l'expression des gènes , Histone/métabolisme , Acétylation , Articulation de la hanche/anatomopathologie , Articulation de la hanche/métabolisme
15.
Life Sci ; 350: 122745, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38834096

RÉSUMÉ

Fibrosis, a pathological state characterized by the excessive accumulation of extracellular matrix components, is primarily driven by the overactivation of fibroblasts. This condition becomes particularly pronounced under chronic inflammatory conditions. Fibrosis can occur in several tissues throughout the body. Among the notable discoveries in the study of fibrosis is the role of Collagen Triple Helix Repeat Containing-1 (CTHRC1), a protein that has emerged as a critical regulator in the fibrotic process. CTHRC1 is rapidly expressed on the outer membrane of fibroblasts and intimal smooth muscle cells following vascular injury, such as that induced by balloon angioplasty. This expression denotes the organism efforts to repair and restructure compromised tissue, signifying a critical component of the tissue repair mechanism in reaction to fibrosis. It plays a pivotal role in promoting cell migration and aiding tissue repair post-injury, contributing significantly to various pathophysiological processes including revascularization, bone formation, developmental morphological changes, inflammatory arthritis, and the progression of cancer. Significantly, researchers have observed marked expression of CTHRC1 across a variety of fibrotic conditions, closely associating it with the progression of the disease. Intervention with CTHRC1 can affect the occurrence and progression of fibrosis. This review aims to comprehensively explore the role and underlying mechanisms of CTHRC1 in fibrotic diseases, highlighting its potential as a key target for therapeutic interventions.


Sujet(s)
Protéines de la matrice extracellulaire , Fibrose , Humains , Fibrose/métabolisme , Protéines de la matrice extracellulaire/métabolisme , Animaux , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Matrice extracellulaire/métabolisme
16.
Arch Dermatol Res ; 316(7): 412, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38878082

RÉSUMÉ

Keloid scars and folliculitis keloidalis nuchae (FKN) are benign fibroproliferative dermal lesions of unknown aetiology and ill-defined treatment, which typically present in genetically susceptible individuals. Their pathognomonic hallmarks include local aggressive invasive behaviour plus high recurrence post-therapy. In view of this, we investigated proliferative and key parameters of bioenergetic cellular characteristics of site-specific keloid-derived fibroblasts (intra(centre)- and peri(margin)-lesional) and FKN compared to normal skin and normal flat non-hypertrophic scar fibroblasts as negative controls.The results showed statistically significant (P < 0.01) and variable growth dynamics with increased proliferation and migration in keloid fibroblasts, while FKN fibroblasts showed a significant (P < 0.001) increase in proliferation but similar migration profile to controls. A statistically significant metabolic switch towards aerobic glycolysis in the fibroblasts from the disease conditions was noted. Furthermore, an increase in basal glycolysis with a concomitant increase in the cellular maximum glycolytic capacity was also demonstrated in perilesional keloid and FKN fibroblasts (P < 0.05). Mitochondrial function parameters showed increased oxidative phosphorylation in the disease conditions (P < 0.05) indicating functional mitochondria. These findings further suggest that Keloids and FKN demonstrate a switch to a metabolic phenotype of aerobic glycolysis. Increased glycolytic flux inhibition is a potential mechanistic basis for future therapy.


Sujet(s)
Prolifération cellulaire , Fibroblastes , Folliculite , Glycolyse , Chéloïde , Humains , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Folliculite/métabolisme , Folliculite/anatomopathologie , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Cellules cultivées , Phosphorylation oxydative , Mouvement cellulaire , Adulte , Peau/anatomopathologie , Peau/métabolisme , Métabolisme énergétique , Femelle , Mâle
17.
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
18.
Respir Res ; 25(1): 242, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38877465

RÉSUMÉ

BACKGROUND: Silicosis represents a paramount occupational health hazard globally, with its incidence, morbidity, and mortality on an upward trajectory, posing substantial clinical dilemmas due to limited effective treatment options available. Trigonelline (Trig), a plant alkaloid extracted mainly from coffee and fenugreek, have diverse biological properties such as protecting dermal fibroblasts against ultraviolet radiation and has the potential to inhibit collagen synthesis. However, it's unclear whether Trig inhibits fibroblast activation to attenuate silicosis-induced pulmonary fibrosis is unclear. METHODS: To evaluate the therapeutic efficacy of Trig in the context of silicosis-related pulmonary fibrosis, a mouse model of silicosis was utilized. The investigation seeks to elucidated Trig's impact on the progression of silica-induced pulmonary fibrosis by evaluating protein expression, mRNA levels and employing Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius Red staining. Subsequently, we explored the mechanism underlying of its functions. RESULTS: In vivo experiment, Trig has been demonstrated the significant efficacy in mitigating SiO2-induced silicosis and BLM-induced pulmonary fibrosis, as evidenced by improved histochemical staining and reduced fibrotic marker expressions. Additionally, we showed that the differentiation of fibroblast to myofibroblast was imped in Trig + SiO2 group. In terms of mechanism, we obtained in vitro evidence that Trig inhibited fibroblast-to-myofibroblast differentiation by repressing TGF-ß/Smad signaling according to the in vitro evidence. Notably, our finding indicated that Trig seemed to be safe in mice and fibroblasts. CONCLUSION: In summary, Trig attenuated the severity of silicosis-related pulmonary fibrosis by alleviating the differentiation of myofibroblasts, indicating the development of novel therapeutic approaches for silicosis fibrosis.


Sujet(s)
Alcaloïdes , Différenciation cellulaire , Fibroblastes , Souris de lignée C57BL , Myofibroblastes , Fibrose pulmonaire , Silice , Silicose , Animaux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Alcaloïdes/pharmacologie , Silice/toxicité , Souris , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Silicose/anatomopathologie , Silicose/métabolisme , Silicose/traitement médicamenteux , Mâle
19.
J Transl Med ; 22(1): 546, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849907

RÉSUMÉ

BACKGROUND: The pathogenesis of thyroid-associated orbitopathy (TAO) remains incompletely understand. The interaction between immunocytes and orbital fibroblasts (OFs) play a critical role in orbital inflammatory and fibrosis. Accumulating reports indicate that a significant portion of plasma exosomes (Pla-Exos) are derived from immune cells; however, their impact upon OFs function is unclear. METHODS: OFs were primary cultured from inactive TAO patients. Exosomes isolated from plasma samples of patients with active TAO and healthy controls (HCs) were utilized for functional and RNA cargo analysis. Functional analysis in thymocyte differentiation antigen-1+ (Thy-1+) OFs measured expression of inflammatory and fibrotic markers (mRNAs and proteins) and cell activity in response to Pla-Exos. RNA cargo analysis was performed by RNA sequencing and RT-qPCR. Thy-1+ OFs were transfected with miR-144-3p mimics/inhibitors to evaluate its regulation of inflammation, fibrosis, and proliferation. RESULTS: Pla-Exos derived from active TAO patients (Pla-ExosTAO-A) induced stronger production of inflammatory cytokines and hyaluronic acid (HA) in Thy-1+ OFs while inhibiting their proliferation. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and single sample gene set enrichment analysis (ssGSEA) suggested that the difference in mRNA expression levels between Pla-ExosTAO-A and Pla-ExosHC was closely related to immune cells. Differential expression analysis revealed that 62 upregulated and 45 downregulated miRNAs in Pla-ExosTAO-A, with the elevation of miR-144-3p in both Pla-Exos and PBMCs in active TAO group. KEGG analysis revealed that the target genes of differentially expressed miRNA and miR-144-3p enriched in immune-related signaling pathways. Overexpression of the miR-144-3p mimic significantly upregulated the secretion of inflammatory cytokines and HA in Thy-1+ OFs while inhibiting their proliferation. CONCLUSION: Pla-Exos derived from patients with active TAO were immune-active, which may be a long-term stimulus casual for inflammatory and fibrotic progression of TAO. Our finding suggests that Pla-Exos could be used as biomarkers or treatment targets in TAO patients.


Sujet(s)
Exosomes , Fibroblastes , Fibrose , Ophtalmopathie basedowienne , Inflammation , microARN , Orbite , Humains , Exosomes/métabolisme , Ophtalmopathie basedowienne/anatomopathologie , Ophtalmopathie basedowienne/sang , Ophtalmopathie basedowienne/génétique , microARN/génétique , microARN/métabolisme , microARN/sang , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Orbite/anatomopathologie , Inflammation/anatomopathologie , Femelle , Mâle , Prolifération cellulaire , Adulte d'âge moyen , Adulte , Acide hyaluronique/sang , Acide hyaluronique/métabolisme , Cytokines/métabolisme , Antigènes Thy-1/métabolisme
20.
Front Biosci (Landmark Ed) ; 29(5): 173, 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38812305

RÉSUMÉ

BACKGROUND: Neointimal hyperplasia (NIH) is the pathological basis of vascular injury disease. Vascular cells are the dominant cells in the process of NIH, but the extent of heterogeneity amongst them is still unclear. METHODS: A mouse model of NIH was constructed by inducing carotid artery ligation. Single-cell sequencing was then used to analyze the transcriptional profile of vascular cells. Cluster features were determined by functional enrichment analysis, gene set scoring, pseudo-time analysis, and cell-cell communication analysis. Additionally, immunofluorescence staining was conducted on vascular tissues from fibroblast lineage-traced (PdgfraDreER-tdTomato) mice to validate the presence of Pecam1+Pdgfra+tdTomato+ cells. RESULTS: The left carotid arteries (ligation) were compared to right carotid arteries (sham) from ligation-induced NIH C57BL/6 mice. Integrative analyses revealed a high level of heterogeneity amongst vascular cells, including fourteen clusters and seven cell types. We focused on three dominant cell types: endothelial cells (ECs), vascular smooth muscle cells (vSMCs), and fibroblasts. The major findings were: (1) four subpopulations of ECs, including ECs4, mesenchymal-like ECs (ECs1 and ECs2), and fibro-like ECs (ECs3); (2) four subpopulations of fibroblasts, including pro-inflammatory Fibs-1, Sca1+ Fibs-2, collagen-producing Fibs-3, and mesenchymal-like Fibs-4; (3) four subpopulations of vSMCs, including vSMCs-1, vSMCs-2, vSMCs-3, and vSMCs-3-derived vSMCs; (4) ECs3 express genes related to extracellular matrix (ECM) remodeling and cell migration, and fibro-like vSMCs showed strong chemokine secretion and relatively high levels of proteases; (5) fibro-like vSMCs that secrete Vegfa interact with ECs mainly through vascular endothelial growth factor receptor 2 (Vegfr2). CONCLUSIONS: This study presents the dynamic cellular landscape within NIH arteries and reveals potential relationships between several clusters, with a specific focus on ECs3 and fibro-like vSMCs. These two subpopulations may represent potential target cells for the treatment of NIH.


Sujet(s)
Analyse de profil d'expression de gènes , Hyperplasie , Souris de lignée C57BL , Muscles lisses vasculaires , Néointima , Analyse sur cellule unique , Animaux , Néointima/anatomopathologie , Néointima/métabolisme , Néointima/génétique , Analyse sur cellule unique/méthodes , Hyperplasie/génétique , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/cytologie , Souris , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Artères carotides/anatomopathologie , Artères carotides/métabolisme , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Mâle , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Modèles animaux de maladie humaine , Analyse de l'expression du gène de la cellule unique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...