Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.664
Filtrer
1.
Front Immunol ; 15: 1373464, 2024.
Article de Anglais | MEDLINE | ID: mdl-39185406

RÉSUMÉ

Introduction: In systemic sclerosis (SSc), B-cells are activated and present in the skin and lung of patients where they can interact with fibroblasts. The precise impact and mechanisms of the interaction of B-cells and fibroblasts at the tissular level are poorly studied. Objective: We investigated the impact and mechanisms of B-cell/fibroblast interactions in cocultures between B-cells from patients with SSc and 3-dimensional reconstituted healthy skin model including fibroblasts, keratinocytes and extracellular matrix. Methods: The quantification and description of the B-cell infiltration in 3D cocultures were performed using cells imagery strategy and cytometry. The effect of coculture on the transcriptome of B-cells and fibroblasts was studied with bulk and single-cell RNA sequencing approaches. The mechanisms of this interaction were studied by blocking key cytokines like IL-6 and TNF. Results: We showed a significant infiltration of B-cells in the 3D healthy skin model. The amount but not the depth of infiltration was higher with B-cells from SSc patients and with activated B-cells. B-cell infiltrates were mainly composed of naïve and memory cells, whose frequencies differed depending on B-cells origin and activation state: infiltrated B-cells from patients with SSc showed an activated profile and an overexpression of immunoglobulin genes compared to circulating B-cells before infiltration. Our study has shown for the first time that activated B-cells modified the transcriptomic profile of both healthy and SSc fibroblasts, toward a pro-inflammatory (TNF and IL-17 signaling) and interferon profile, with a key role of the TNF pathway. Conclusion: B-cells and 3D skin cocultures allowed the modelization of B-cells infiltration in tissues observed in SSc, uncovering an influence of the underlying disease and the activation state of B-cells. We showed a pro-inflammatory effect on skin fibroblasts and pro-activation effect on infiltrating B-cells during coculture. This reinforces the role of B-cells in SSc and provide potential targets for future therapeutic approach in this disease.


Sujet(s)
Lymphocytes B , Techniques de coculture , Fibroblastes , Sclérodermie systémique , Peau , Humains , Sclérodermie systémique/immunologie , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme , Fibroblastes/immunologie , Fibroblastes/métabolisme , Peau/immunologie , Peau/anatomopathologie , Peau/métabolisme , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Femelle , Communication cellulaire/immunologie , Activation des lymphocytes/immunologie , Adulte d'âge moyen , Mâle , Cellules cultivées , Transcriptome , Adulte , Kératinocytes/immunologie , Kératinocytes/métabolisme , Cytokines/métabolisme
2.
Dokl Biochem Biophys ; 517(1): 228-234, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39002011

RÉSUMÉ

The pathogenesis of immunoinflammatory rheumatic diseases (IRDs) is based on chronic inflammation, one of the key mechanisms of which may be abnormal activation of macrophages, leading to further disruption of the immune system. OBJECTIVE: . The objective of this study was to evaluate the proinflammatory activation of circulating monocytes in patients with IRDs. MATERIALS AND METHODS: . The study involved 149 participants (53 patients with rheumatoid arthritis (RA), 45 patients with systemic lupus erythematosus (SLE), 34 patients with systemic scleroderma (SSc), and 17 participants without IRDs) 30 to 65 years old. Basal and lipopolysaccharide (LPS)-stimulated secretion of monocytes was studied in a primary culture of monocytes obtained from blood by immunomagnetic separation. Quantitative assessment of the cytokines tumor necrosis factor α (TNF-α), interleukin 1ß (IL-1ß), as well as the chemokine monocyte chemoattractant protein-1 (MCP-1) was carried out in the culture fluid by ELISA. Proinflammatory activation of monocytes was calculated as the ratio of LPS-stimulated and basal secretions. RESULTS: . It was shown that the basal secretion of all studied cytokines was significantly increased in all groups of patients with IRDs, except for the secretion of IL-1ß in the SLE group, compared to the control. LPS-stimulated secretion of TNF-α was increased and MCP-1 was decreased in patients with IRDs compared to the control group; LPS-stimulated IL-1ß secretion only in the SSc group significantly differed from the control group. In the RA group, monocyte activation was reduced for all cytokines compared to the control; in the SLE group, for TNF-α and MCP-1; in the SSc group, for MCP-1. CONCLUSIONS: . The decrease in proinflammatory activation of monocytes in patients with IRDs is due to a high level of basal secretion of cytokines, which can lead to disruption of the adequate immune response in these diseases and is an important link in the pathogenesis of chronic inflammation.


Sujet(s)
Inflammation , Monocytes , Humains , Monocytes/immunologie , Monocytes/métabolisme , Adulte d'âge moyen , Adulte , Femelle , Mâle , Inflammation/immunologie , Inflammation/métabolisme , Lipopolysaccharides/pharmacologie , Sujet âgé , Chimiokine CCL2/métabolisme , Polyarthrite rhumatoïde/immunologie , Rhumatismes/immunologie , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-1 bêta/métabolisme , Sclérodermie systémique/immunologie , Sclérodermie systémique/métabolisme , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , Cytokines/métabolisme
3.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000334

RÉSUMÉ

Systemic sclerosis (SSc) is characterized by dermal fibrosis with a female predominance, suggesting a hormonal influence. Patients with SSc have elevated interleukin (IL)-6 levels, and post-menopausal women and older men also have high estradiol (E2) levels. In the skin, IL-6 increases the enzymatic activity of aromatase, thereby amplifying the conversion of testosterone to E2. Therefore, we hypothesized that an interplay between E2 and IL-6 contributes to dermal fibrosis. We used primary dermal fibroblasts from healthy donors and patients with diffuse cutaneous (dc)SSc, and healthy donor skin tissues stimulated with recombinant IL-6 and its soluble receptor (sIL-6R) or E2. Primary human dermal fibroblasts and tissues from healthy donors stimulated with IL-6+sIL-6R produced E2, while E2-stimulated dermal tissues and fibroblasts produced IL-6. Primary dermal fibroblasts from healthy donors treated with IL-6+sIL-6R and the aromatase inhibitor anastrozole (ANA) and dcSSc fibroblasts treated with ANA produced less fibronectin (FN), type III collagen A1 (Col IIIA1), and type V collagen A1 (Col VA1). Finally, dcSSc dermal fibroblasts treated with the estrogen receptor inhibitor fulvestrant also generated less FN, Col IIIA1, and Col VA1. Our data show that IL-6 exerts its pro-fibrotic influence in human skin in part through E2 and establish a positive feedback loop between E2 and IL-6.


Sujet(s)
Oestradiol , Fibroblastes , Fibrose , Interleukine-6 , Sclérodermie systémique , Humains , Interleukine-6/métabolisme , Oestradiol/pharmacologie , Oestradiol/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/anatomopathologie , Femelle , Mâle , Peau/métabolisme , Peau/anatomopathologie , Cellules cultivées , Rétrocontrôle physiologique , Adulte d'âge moyen , Adulte , Récepteurs à l'interleukine-6/métabolisme
4.
Scand J Immunol ; 99(4): e13354, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-39008522

RÉSUMÉ

Systemic sclerosis (SSc) is a chronic autoimmune connective tissue disease. Vascular damage is one of the important features of SSc, which affects the progression and prognosis of the disease. MiR-126-3p is an important microRNA (miRNA) that regulates vascular structure and function, which can be transported through exosomes. However, the role of miR-126-3p in vascular damage in SSc is still unclear. Therefore, we focused on the connection between miR-126-3p and vascular damage in SSc, as well as investigated the potential role of miR-126-3p in vascular damage in SSc. First, this study successfully extracted extracellular vesicles from clinical plasma samples and characterized the exosomes within them. Then, we predicted and screened the target pathway mammalian/mechanistic target of rapamycin (mTOR) and the target gene SLC7A5 of miR-126-3p through online databases. Next, we constructed SSc mice for in vivo studies. The results showed that the expression of miR-126-3p was decreased in the plasma exosomes, while the SLC7A5 expression, autophagy, and lipid peroxidation were increased in the aorta. Luciferase reporter gene assays demonstrated that miR-126-3p can bind to SLC7A5, resulting in a decrease in its expression. In vitro experiments have shown that exosomal miR-126-3p can be internalized by human umbilical vein endothelial cells (HUVECs). The miR-126-3p group exhibited enhanced cell viability and tube formation ability, along with increased expression of the vascular formation marker CD31. Additionally, miR-126-3p downregulated the protein expression of SLC7A5 and LC3 in HUVECs, while upregulating the protein expression of mTOR, P62, PPARγ, and CPT-1. However, the effects of miR-126-3p on HUVECs were counteracted by mTOR inhibitors and enhanced by mTOR activators. The results indicated that exosomal miR-126-3p has the potential to protect against vascular injury in SSc by regulating the SLC7A5/mTOR signalling pathway in HUVECs.


Sujet(s)
Exosomes , Cellules endothéliales de la veine ombilicale humaine , microARN , Transduction du signal , Sérine-thréonine kinases TOR , microARN/génétique , microARN/métabolisme , Humains , Exosomes/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Animaux , Souris , Sclérodermie systémique/métabolisme , Sclérodermie systémique/génétique , Sclérodermie systémique/anatomopathologie , Mâle , Femelle , Transporteur-1 d'acides aminés neutres à longue chaîne/métabolisme , Transporteur-1 d'acides aminés neutres à longue chaîne/génétique , Adulte d'âge moyen , Modèles animaux de maladie humaine , Adulte
5.
Front Biosci (Landmark Ed) ; 29(7): 259, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39082361

RÉSUMÉ

BACKGROUND: Investigation of the inflammatory response of immune cells is a current focus of research on autoimmune disorders. The aim of this study was to evaluate the inflammatory status of monocytes/macrophages in systemic sclerosis (SSc). METHODS: The study included 35 SSc and 25 healthy participants. The secretion of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), monocyte chemoattractant protein-1 (MCP-1), interleukin-8 (IL-8), interleukin-6 (IL-6) was measured by enzyme-linked immunosorbent assay (ELISA) in primary cultures of monocytes/macrophages after stimulation with lipopolysaccharide (LPS) on day 1 and on day 6 of incubation. Impaired tolerance of the immune response was characterized by increased secretion of the inflammatory mediators in response to restimulation. RESULTS: Basal secretion of all cytokines was significantly higher in SSc patients compared to healthy individuals. The secretion of TNF-α, IL-1ß and IL-6 after the initial LPS stimulation, and secretion of IL-1ß, MCP-1, IL-6, IL-8 after LPS restimulation, was significantly higher in the SSc group. Eleven SSc patients (31%) showed impaired immune tolerance in terms of MCP-1 secretion. These patients were significantly younger and had a higher level of anti-topoisomerase I (anti-Scl70) antibodies compared to SSc patients with immune tolerance. CONCLUSIONS: This study revealed pro-inflammatory activation and impaired immune tolerance in monocytes/macrophages from SSc patients. The violation of immune response in terms of MCP-1 secretion may be an important factor in the development of chronic inflammation in SSc. MCP-1 may thus be a potential therapeutic target for novel SSc treatment strategies.


Sujet(s)
Macrophages , Monocytes , Sclérodermie systémique , Humains , Sclérodermie systémique/immunologie , Sclérodermie systémique/métabolisme , Monocytes/immunologie , Monocytes/métabolisme , Femelle , Mâle , Adulte d'âge moyen , Macrophages/immunologie , Macrophages/métabolisme , Adulte , Inflammation/immunologie , Lipopolysaccharides , Cytokines/métabolisme , Cytokines/immunologie , Études cas-témoins , Chimiokine CCL2/métabolisme , Chimiokine CCL2/immunologie , Sujet âgé , Test ELISA , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/immunologie
6.
Cells ; 13(14)2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39056787

RÉSUMÉ

Systemic sclerosis (SSc) is an autoimmune connective tissue disease with a triad of features that include vascular abnormalities, inflammation and skin and lung fibrosis. At the core of the disease is the activation of myofibroblasts from quiescent fibroblasts and this can be modified by various cytokines. IL-41 is a recently described cytokine that was initially characterised as an adipokine as it was highly expressed in adipocytes and adipose tissue. However, it has recently been identified as being widely expressed and has immunomodulatory functions. This study examined the circulating levels of IL-41 and its expression in skin biopsies. We demonstrated significantly reduced levels of IL-41 in diffuse SSc that was also mirrored in the skin of SSc patients. AMPK has been proposed as a downstream target of IL-41, so we also measure mammalian target of rapamycin in skin and found that this is elevated in SSc patients. We speculate that IL-41 maybe an antifibrotic cytokine and its reduction may facilitate the activation of fibroblasts.


Sujet(s)
Interleukines , Peau , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Cytokines/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Interleukines/métabolisme , Sclérodermie diffuse/anatomopathologie , Sclérodermie diffuse/métabolisme , Sclérodermie diffuse/sang , Sclérodermie diffuse/génétique , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/sang , Peau/anatomopathologie , Peau/métabolisme , Sérine-thréonine kinases TOR/métabolisme
7.
Clin Exp Rheumatol ; 42(8): 1623-1628, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38976307

RÉSUMÉ

OBJECTIVES: BAG3 (Bcl2-associated athanogene3) is able to induce the transformation of cancer-associated fibroblasts to alpha smooth muscle actin (a-SMA) positive (+) myofibroblasts. In systemic sclerosis (SSc), a-SMA+ myofibroblasts also play an important role in the progression of fibrosis in the skin and involved internal organs. The aim of the study was to investigate whether BAG3 is overexpressed in SSc and may be a biomarker of fibrogenesis. METHODS: BAG3 serum levels were measured in 106 patients with SSc, 47 with the limited (lc) and 59 the diffuse (dc) SSc, and in age- and sex-matched healthy controls (HC). BAG3 levels were then compared according to their clinical subset, nailfold video-capillaroscopic (NVC) patterns, interstitial lung disease (ILD, and correlated with modified Rodnan skin score (mRSS) and global disease activity. BAG3 expression was also investigated in skin biopsies of 8 dcSSc patients. RESULTS: BAG3 serum levels were significantly higher in dcSSc (143.3 pg/mL, 95%CI 78-208.5) than in HC (0.68 pg/mL, 95%CI 0.13-1.23), and were significantly higher in patients with late NVC pattern and ILD but did not correlate with disease activity and mRSS. Of note, BAG3 was strongly expressed in the skin biopsies of dcSSc patients. CONCLUSIONS: BAG3 is overexpressed in dcSSc patients and may contribute to skin and organ fibrosis by prompting the transition of fibroblasts into myofibroblasts and increasing their survival. Thus, BAG3 may play an important role in SSc fibrotic pathogenesis and be a potential biomarker of fibrosis. Further research on its role as a therapeutic target is warranted.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines régulatrices de l'apoptose , Marqueurs biologiques , Peau , Humains , Femelle , Mâle , Adulte d'âge moyen , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/sang , Peau/anatomopathologie , Peau/métabolisme , Adulte , Marqueurs biologiques/sang , Études cas-témoins , Fibrose , Sujet âgé , Régulation positive , Sclérodermie diffuse/sang , Sclérodermie limitée/sang , Sclérodermie limitée/diagnostic , Biopsie , Sclérodermie systémique/sang , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme
8.
Front Immunol ; 15: 1400819, 2024.
Article de Anglais | MEDLINE | ID: mdl-38863696

RÉSUMÉ

Background: Integrin-dependent cell adhesion and migration play important roles in systemic sclerosis (SSc). The roles of integrin activating molecules including talins and kindlins, however, are unclear in SSc. Objectives: We aimed to explore the function of integrin activating molecules in SSc. Methods: Transcriptome analysis of skin datasets of SSc patients was performed to explore the function of integrin-activating molecules including talin1, talin2, kindlin1, kindlin2 and kindlin3 in SSc. Expression of talin1 in skin tissue was assessed by multiplex immunohistochemistry staining. Levels of talin1 in serum were determined by ELISA. The effects of talin1 inhibition were analyzed in human dermal fibroblasts by real-time PCR, western blot and flow cytometry. Results: We identified that talin1 appeared to be the primary integrin activating molecule involved in skin fibrosis of SSc. Talin1 was significantly upregulated and positively correlates with the modified Rodnan skin thickness score (mRSS) and the expression of pro-fibrotic biomarkers in the skin lesions of SSc patients. Further analyses revealed that talin1 is predominantly expressed in the dermal fibroblasts of SSc skin and promotes fibroblast activation and collagen production. Additionally, talin1 primarily exerts its effects through integrin ß1 and ß5 in SSc. Conclusions: Overexpressed talin1 is participated in skin fibrosis of SSc, and talin1 appears to be a potential new therapeutic target for SSc.


Sujet(s)
Fibroblastes , Fibrose , Sclérodermie systémique , Peau , Taline , Humains , Sclérodermie systémique/métabolisme , Sclérodermie systémique/anatomopathologie , Taline/métabolisme , Taline/génétique , Peau/métabolisme , Peau/anatomopathologie , Fibroblastes/métabolisme , Femelle , Mâle , Adulte d'âge moyen , Adulte , Cellules cultivées
9.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38892317

RÉSUMÉ

The bleomycin-induced scleroderma model is a well-established and dependable method for creating a mouse model of SSc (systemic sclerosis). In the field of skin connective tissue diseases, increasing evidence from clinical and animal experiments suggests that TLRs (Toll-like receptors) play an important role in several diseases. This study aimed to determine the role of TLR7 (Toll-like receptor 7) and TLR9 (Toll-like receptor 9) in the mechanisms of immune abnormalities and fibrosis in SSc. This study used TLR7-KO mice (TLR7-knockout mice with a balb/c background) and TLR9-KO mice (TLR9-knockout mice with a balb/c background) as well as WT mice (wild-type balb/c mice). All three kinds of mice were induced by BLM (bleomycin) in a scleroderma model as the experimental group; meanwhile, WT mice treated with PBS (phosphate-buffered saline) were used as the control group. We analyzed the fibrotic phenotype and the immunological abnormality phenotype of TLR7-deficient and TLR9-deficient mice in the SSc disease model using flow cytometry, RT-PCR (reverse transcription-polymerase chain reaction), a histological examination, and IHC (immunohistochemical staining). In a mouse model of SSc disease, the deletion of TLR7 attenuated skin and lung fibrosis, while the deletion of TLR9 exacerbated skin and lung fibrosis. The deletion of TLR7 resulted in a relative decrease in the infiltration and expression of various pro-inflammatory and fibrotic cells and cytokines in the skin. On the other hand, the deletion of TLR9 resulted in a relative increase in the infiltration and expression of various pro-inflammatory and cytokine-inhibiting cells and cytokines in the skin. Under the influence of pDCs (plasmacytoid dendritic cells), the balances of Beff/Breg (IL-6 + CD19 + B cell/IL-10 + CD19 + B cell), Th17/Treg (IL-17A + CD4 + T cell/Foxp3 + CD25 + CD4 + T cell), M1/M2 (CD86 + macrophage/CD206 + macrophage), and Th1/Th2 (TNFα + CD3 + CD4 + T cell/IL-4 + CD3 + CD4 + T cell) were biased towards the suppression of inflammation and fibrosis as a result of the TLR7 deletion. Comparatively, the balance was biased towards promoting inflammation and fibrosis due to the TLR9 deletion. In the SSc model, TLR7 promoted inflammation and fibrosis progression, while TLR9 played a protective role. These results suggest that TLR7 and TLR9 play opposite roles in triggering SSc to produce immune system abnormalities and skin fibrosis.


Sujet(s)
Modèles animaux de maladie humaine , Souris knockout , Sclérodermie systémique , Récepteur de type Toll-7 , Récepteur-9 de type Toll-like , Animaux , Récepteur de type Toll-7/métabolisme , Récepteur de type Toll-7/génétique , Sclérodermie systémique/métabolisme , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/immunologie , Sclérodermie systémique/génétique , Récepteur-9 de type Toll-like/métabolisme , Récepteur-9 de type Toll-like/génétique , Souris , Bléomycine/effets indésirables , Souris de lignée BALB C , Cytokines/métabolisme , Peau/anatomopathologie , Peau/métabolisme , Peau/immunologie , Fibrose , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/étiologie , Glycoprotéines membranaires
10.
Arterioscler Thromb Vasc Biol ; 44(8): e210-e225, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38841857

RÉSUMÉ

BACKGROUND: Pulmonary hypertension (PH) is a common complication of systemic sclerosis (SSc) and a leading cause of mortality among patients with this disease. PH can also occur as an idiopathic condition (idiopathic pulmonary arterial hypertension). Investigation of transcriptomic alterations in vascular populations is critical to elucidating cellular mechanisms underlying pathobiology of SSc-associated and idiopathic PH. METHODS: We analyzed single-cell RNA sequencing profiles of endothelial and perivascular mesenchymal populations from explanted lung tissue of patients with SSc-associated PH (n=16), idiopathic pulmonary arterial hypertension (n=3), and healthy controls (n=15). Findings were validated by immunofluorescence staining of explanted human lung tissue. RESULTS: Three disease-associated endothelial populations emerged. Two angiogenic endothelial cell (EC) subtypes markedly expanded in SSc-associated PH lungs: tip ECs expressing canonical tip markers PGF and APLN and phalanx ECs expressing genes associated with vascular development, endothelial barrier integrity, and Notch signaling. Gene regulatory network analysis suggested enrichment of Smad1 (SMAD family member 1) and PPAR-γ (peroxisome proliferator-activated receptor-γ) regulon activities in these 2 populations, respectively. Mapping of potential ligand-receptor interactions highlighted Notch, apelin-APJ (apelin receptor), and angiopoietin-Tie (tyrosine kinase with immunoglobulin-like and EGF-like domains 1) signaling pathways between angiogenic ECs and perivascular cells. Transitional cells, expressing both endothelial and pericyte/smooth muscle cell markers, provided evidence for the presence of endothelial-to-mesenchymal transition. Transcriptional programs associated with arterial endothelial dysfunction implicated VEGF-A (vascular endothelial growth factor-A), TGF-ß1 (transforming growth factor beta-1), angiotensin, and TNFSF12 (tumor necrosis factor ligand superfamily member 12)/TWEAK (TNF-related weak inducer of apoptosis) in the injury/remodeling phenotype of PH arterial ECs. CONCLUSIONS: These data provide high-resolution insights into the complexity and plasticity of the pulmonary endothelium in SSc-associated PH and idiopathic pulmonary arterial hypertension and provide direct molecular insights into soluble mediators and transcription factors driving PH vasculopathy.


Sujet(s)
Néovascularisation pathologique , Sclérodermie systémique , Remodelage vasculaire , Humains , Sclérodermie systémique/génétique , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/complications , Mâle , Femelle , Adulte d'âge moyen , Hypertension artérielle pulmonaire primitive familiale/métabolisme , Hypertension artérielle pulmonaire primitive familiale/génétique , Hypertension artérielle pulmonaire primitive familiale/physiopathologie , Hypertension artérielle pulmonaire primitive familiale/anatomopathologie , Études cas-témoins , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Artère pulmonaire/physiopathologie , Transcriptome , Transduction du signal , Adulte , Analyse sur cellule unique , Poumon/métabolisme , Poumon/vascularisation , Poumon/anatomopathologie , Réseaux de régulation génique ,
11.
Matrix Biol ; 132: 24-33, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38852924

RÉSUMÉ

Pulmonary fibrosis (PF) is a clinically severe and commonly fatal complication of Systemic Sclerosis (SSc). Our group has previously reported profibrotic roles for Insulin-like Growth Factor II (IGF-II) and Lysyl Oxidase (LOX) in SSc-PF. We sought to identify downstream regulatory mediators of IGF-II. In the present work, we show that SSc lung tissues have higher baseline levels of the total (N-glycosylated/unglycosylated) LOX-Propeptide (LOX-PP) than control lung tissues. LOX-PP-mediated changes were consistent with the extracellular matrix (ECM) deregulation implicated in SSc-PF progression. Furthermore, Tolloid-like 1 (TLL1) and Bone Morphogenetic Protein 1 (BMP1), enzymes that can cleave ProLOX to release LOX-PP, were increased in SSc lung fibrosis and the bleomycin (BLM)-induced murine lung fibrosis model, respectively. In addition, IGF-II regulated the levels of ProLOX, active LOX, LOX-PP, BMP1, and isoforms of TLL1. The Class E Basic Helix-Loop-Helix protein 40 (BHLHE40) transcription factor localized to the nucleus in response to IGF-II. BHLHE40 silencing downregulated TLL1 isoforms and LOX-PP, and restored features of ECM deregulation triggered by IGF-II. Our findings indicate that IGF-II, BHLHE40, and LOX-PP may serve as targets of therapeutic intervention to halt SSc-PF progression.


Sujet(s)
Protéine morphogénétique osseuse de type 1 , Facteur de croissance IGF-II , Lysyloxidase , Fibrose pulmonaire , Animaux , Lysyloxidase/métabolisme , Lysyloxidase/génétique , Facteur de croissance IGF-II/métabolisme , Facteur de croissance IGF-II/génétique , Souris , Humains , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/génétique , Protéine morphogénétique osseuse de type 1/métabolisme , Protéine morphogénétique osseuse de type 1/génétique , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Matrice extracellulaire/métabolisme , Poumon/métabolisme , Poumon/anatomopathologie , Bléomycine/pharmacologie , Modèles animaux de maladie humaine , Femelle , Mâle
12.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 53(3): 376-381, 2024 Jun 19.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38899353

RÉSUMÉ

The core components of the Hippo signaling pathway encompass upstream regulatory molecules, core kinase cascade complexes, and downstream transcriptional regulation complexes. This pathway modulates cellular behaviors by influencing the effector molecules of its core components and plays a pivotal role in immune regulation. Effector molecules,such as Yes-associated protein (YAP), transcriptional coactivator with PDZ-binding motif (TAZ), transcriptional enhanced associate domain transcriptional factor (TEAD), monopolar spindle-one binder (MOB1), large tumor suppressor (LATS), can stimulate fibroblast-like synovial cell migration and invasion in rheumatoid arthritis, regulate osteoarthritis disease progression, promote pathological new bone formation in ankylosing spondylitis, sustain submandibular gland development while delaying Sjogren's syndrome progression, mediate alpha-smooth muscle actin in systemic sclerosis, and refine the regulation of target genes associated with pulmonary fibrosis. This article provides an overview of the regulatory mechanisms involving Hippo signaling pathway-related effector molecules in the pathogenesis and progression of rheumatic immune system diseases, to serve as a reference for exploring novel therapeutic targets of rheumatic immune system diseases.


Sujet(s)
Voie de signalisation Hippo , Protein-Serine-Threonine Kinases , Transduction du signal , Facteurs de transcription , Humains , Protein-Serine-Threonine Kinases/métabolisme , Facteurs de transcription/métabolisme , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/métabolisme , Rhumatismes/immunologie , Rhumatismes/métabolisme , Sclérodermie systémique/métabolisme , Sclérodermie systémique/immunologie , Syndrome de Gougerot-Sjögren/métabolisme , Syndrome de Gougerot-Sjögren/immunologie , Pelvispondylite rhumatismale/immunologie , Pelvispondylite rhumatismale/métabolisme , Maladies du système immunitaire/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme
13.
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
14.
J Clin Invest ; 134(10)2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38747285

RÉSUMÉ

Transforming growth factor ß (TGF-ß) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-ß remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-ß in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-ß. The activation of latent TGF-ß requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-ß, rebalanced TGF-ß signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-ß in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.


Sujet(s)
Fibroblastes , Fibrose , Facteur de croissance transformant bêta , Protéine Wnt-5a , rho-Associated Kinases , Protéine Wnt-5a/métabolisme , Protéine Wnt-5a/génétique , Animaux , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Souris , Humains , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/génétique , Souris knockout , Protéines de type Wingless/métabolisme , Protéines de type Wingless/génétique , Système de signalisation des MAP kinases , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Transduction du signal , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/génétique
15.
Exp Dermatol ; 33(5): e15083, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38794808

RÉSUMÉ

Interferons (IFNs) are signalling proteins primarily involved in initiating innate immune responses against pathogens and promoting the maturation of immune cells. Interferon Regulatory Factor 7 (IRF7) plays a pivotal role in the IFNs signalling pathway. The activation process of IRF7 is incited by exogenous or abnormal nucleic acids, which is followed by the identification via pattern recognition receptors (PRRs) and the ensuing signalling cascades. Upon activation, IRF7 modulates the expression of both IFNs and inflammatory gene regulation. As a multifunctional transcription factor, IRF7 is mainly expressed in immune cells, yet its presence is also detected in keratinocytes, fibroblasts, and various dermal cell types. In these cells, IRF7 is critical for skin immunity, inflammation, and fibrosis. IRF7 dysregulation may lead to autoimmune and inflammatory skin conditions, including systemic scleroderma (SSc), systemic lupus erythematosus (SLE), Atopic dermatitis (AD) and Psoriasis. This comprehensive review aims to extensively elucidate the role of IRF7 and its signalling pathways in immune cells and keratinocytes, highlighting its significance in skin-related and connective tissue diseases.


Sujet(s)
Maladies du tissu conjonctif , Facteur-7 de régulation d'interféron , Kératinocytes , Transduction du signal , Maladies de la peau , Humains , Facteur-7 de régulation d'interféron/métabolisme , Facteur-7 de régulation d'interféron/génétique , Maladies de la peau/immunologie , Maladies de la peau/métabolisme , Kératinocytes/métabolisme , Kératinocytes/immunologie , Maladies du tissu conjonctif/métabolisme , Maladies du tissu conjonctif/immunologie , Psoriasis/immunologie , Psoriasis/métabolisme , Animaux , Peau/métabolisme , Peau/immunologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/immunologie , Sclérodermie systémique/génétique , Immunité innée
16.
Int J Rheum Dis ; 27(5): e15174, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38720423

RÉSUMÉ

OBJECTIVES: This study investigates the role of TNF-induced protein 3 (TNFAIP3) and CCAAT/enhancer-binding protein ß (C/EBPß) in alveolar macrophages (AMs) of patients with systemic sclerosis-associated interstitial lung disease (SSc-ILD) and their influence on pulmonary fibrosis. METHODS: Transfection of HEK293T cells and AMs with plasmids carrying TNFAIP3 and C/EBPß was performed, followed by co-culturing AMs with pulmonary fibroblasts. Immunoblotting analysis was then utilized to assess the expression of TNFAIP3, C/EBPß, and collagen type 1 (Col1). Quantitative PCR analysis was conducted to quantify the mRNA levels of C/EBPß, IL-10, and TGF-ß1. STRING database analysis, and immunoprecipitation assays were employed to investigate the interactions between TNFAIP3 and C/EBPß. RESULTS: TNFAIP3 expression was significantly reduced in SSc-ILD AMs, correlating with increased Col1 production in fibroblasts. Overexpression of TNFAIP3 inhibited this pro-fibrotic activity. Conversely, C/EBPß expression was elevated in SSc-ILD AMs, and its reduction through TNFAIP3 restoration decreased pro-fibrotic cytokines IL-10 and TGFß1 levels. Protein-protein interaction studies confirmed the regulatory relationship between TNFAIP3 and C/EBPß. CONCLUSIONS: This study highlights the important role of TNFAIP3 in regulating pulmonary fibrosis in SSc-ILD by modulating C/EBPß expression in AMs. These findings suggest that targeting TNFAIP3 could be a potential therapeutic strategy for managing SSc-ILD patients.


Sujet(s)
Protéine bêta de liaison aux séquences stimulatrices de type CCAAT , Techniques de coculture , Fibroblastes , Pneumopathies interstitielles , Macrophages alvéolaires , Sclérodermie systémique , Protéine-3 induite par le facteur de nécrose tumorale alpha , Femelle , Humains , Mâle , Adulte d'âge moyen , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Collagène de type I/métabolisme , Collagène de type I/génétique , Fibroblastes/métabolisme , Cellules HEK293 , Interleukine-10/métabolisme , Interleukine-10/génétique , Poumon/métabolisme , Poumon/anatomopathologie , Pneumopathies interstitielles/métabolisme , Pneumopathies interstitielles/étiologie , Macrophages alvéolaires/métabolisme , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/étiologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/complications , Transduction du signal , Facteur de croissance transformant bêta-1/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/génétique , Adulte , Sujet âgé
17.
J Autoimmun ; 146: 103219, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38696927

RÉSUMÉ

Tissue repair is disturbed in fibrotic diseases like systemic sclerosis (SSc), where the deposition of large amounts of extracellular matrix components such as collagen interferes with organ function. LAIR-1 is an inhibitory collagen receptor highly expressed on tissue immune cells. We questioned whether in SSc, impaired LAIR-1-collagen interaction is contributing to the ongoing inflammation and fibrosis. We found that SSc patients do not have an intrinsic defect in LAIR-1 expression or function. Instead, fibroblasts from healthy controls and SSc patients stimulated by soluble factors that drive inflammation and fibrosis in SSc deposit disorganized collagen products in vitro, which are dysfunctional LAIR-1 ligands. This is dependent of matrix metalloproteinases and platelet-derived growth factor receptor signaling. In support of a non-redundant role of LAIR-1 in the control of fibrosis, we found that LAIR-1-deficient mice have increased skin fibrosis in response to repeated injury and in the bleomycin mouse model for SSc. Thus, LAIR-1 represents an essential control mechanism for tissue repair. In fibrotic disease, excessive collagen degradation may lead to a disturbed feedback loop. The presence of functional LAIR-1 in patients provides a therapeutic opportunity to reactivate this intrinsic negative feedback mechanism in fibrotic diseases.


Sujet(s)
Collagène , Modèles animaux de maladie humaine , Fibroblastes , Fibrose , Souris knockout , Récepteurs immunologiques , Sclérodermie systémique , Animaux , Humains , Sclérodermie systémique/immunologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/anatomopathologie , Souris , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Collagène/métabolisme , Fibroblastes/métabolisme , Bléomycine/effets indésirables , Peau/anatomopathologie , Peau/métabolisme , Peau/immunologie , Transduction du signal , Mâle , Femelle , Cellules cultivées
18.
Environ Pollut ; 355: 124194, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38782158

RÉSUMÉ

Nowadays, silica products are widely used in daily life, especially in skin applications, which inevitably increases the risk of silica exposure in general population. However, inadequate awareness of silica's potential hazards and lack of self-protection are of concern. Systemic sclerosis (SSc) is characterized by progressive tissue fibrosis under environmental and genetic interactions. Silica exposure is considered an important causative factor for SSc, but its pathogenesis remains unclear. Within this study, we showed that lower doses of silica significantly promoted the proliferation, migration, and activation of human skin fibroblasts (HSFs) within 24 h. Silica injected subcutaneously into mice induced and exacerbated skin fibrosis. Notably, silica increased histone deacetylase-4 (HDAC4) expression by inducing its DNA hypomethylation in normal HSFs. The elevated HDAC4 expression was also confirmed in SSc HSFs. Furthermore, HDAC4 was positively correlated with Smad2/3 phosphorylation and COL1, α-SMA, and CTGF expression. The HDAC4 inhibitor LMK235 mitigated silica-induced upregulation of these factors and alleviated skin fibrosis in SSc mice. Taken together, silica induces and exacerbates skin fibrosis in SSc patients by targeting the HDAC4/Smad2/3 pathway. Our findings provide new insights for evaluating the health hazards of silica exposure and identify HDAC4 as a potential interventional target for silica-induced SSc skin fibrosis.


Sujet(s)
Fibrose , Histone deacetylases , Sclérodermie systémique , Silice , Peau , Protéine Smad2 , Protéine Smad-3 , Sclérodermie systémique/métabolisme , Sclérodermie systémique/induit chimiquement , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Animaux , Souris , Humains , Protéine Smad-3/métabolisme , Peau/métabolisme , Protéine Smad2/métabolisme , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Protéines de répression/métabolisme , Protéines de répression/génétique , Transduction du signal/effets des médicaments et des substances chimiques
19.
Free Radic Biol Med ; 221: 23-30, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38740100

RÉSUMÉ

The pathogenesis of systemic sclerosis (SSC) fibrosis involves the rapid proliferation of skin fibroblasts, and current anti-fibrotic treatments are limited. This study investigated the relationship between ferroptosis and SSC skin fibroblasts. We observed that erastin-induced ferroptosis was suppressed in SSC fibroblasts. RSL3, a direct inhibitor of Glutathione Peroxidase 4 (GPX4), significantly reduced the viability of the fibroblasts, and upregulation of GPX4 in the SSC fibroblasts contributed to ferroptosis resistance. Furthermore, we demonstrated that transferrin receptor 1 (TfR1) was a crucial transporter for iron deposition in the fibroblasts. Collectively, our results highlight that GPX4 inhibition could enhance the sensitivity to ferroptosis by SSC fibroblasts, which showed distinct characteristics of iron metabolism that were not observed in normal fibroblasts in this study. Taken together, these results suggest that targeting ferroptosis could be a therapeutic strategy for the treatment of SSC.


Sujet(s)
Ferroptose , Fibroblastes , Phospholipid hydroperoxide glutathione peroxidase , Sclérodermie systémique , Peau , Femelle , Humains , Antigènes CD/métabolisme , Antigènes CD/génétique , Carbolines , Cellules cultivées , Ferroptose/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/effets des médicaments et des substances chimiques , Fer/métabolisme , Phénanthridines/pharmacologie , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Pipérazines , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/métabolisme , Sclérodermie systémique/génétique , Peau/anatomopathologie , Peau/métabolisme , Régulation positive
20.
JCI Insight ; 9(10)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38652537

RÉSUMÉ

NKX2-5 is a member of the homeobox-containing transcription factors critical in regulating tissue differentiation in development. Here, we report a role for NKX2-5 in vascular smooth muscle cell phenotypic modulation in vitro and in vascular remodeling in vivo. NKX2-5 is upregulated in scleroderma patients with pulmonary arterial hypertension. Suppression of NKX2-5 expression in smooth muscle cells halted vascular smooth muscle proliferation and migration, enhanced contractility, and blocked the expression of extracellular matrix genes. Conversely, overexpression of NKX2-5 suppressed the expression of contractile genes (ACTA2, TAGLN, CNN1) and enhanced the expression of matrix genes (COL1) in vascular smooth muscle cells. In vivo, conditional deletion of NKX2-5 attenuated blood vessel remodeling and halted the progression to hypertension in a mouse chronic hypoxia model. This study revealed that signals related to injury such as serum and low confluence, which induce NKX2-5 expression in cultured cells, is potentiated by TGF-ß and further enhanced by hypoxia. The effect of TGF-ß was sensitive to ERK5 and PI3K inhibition. Our data suggest a pivotal role for NKX2-5 in the phenotypic modulation of smooth muscle cells during pathological vascular remodeling and provide proof of concept for therapeutic targeting of NKX2-5 in vasculopathies.


Sujet(s)
Protéine homéotique Nkx-2.5 , Muscles lisses vasculaires , Remodelage vasculaire , Animaux , Souris , Protéine homéotique Nkx-2.5/génétique , Protéine homéotique Nkx-2.5/métabolisme , Humains , Remodelage vasculaire/génétique , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Mâle , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/complications , Sclérodermie systémique/métabolisme , Sclérodermie systémique/génétique , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/génétique , Hypertension artérielle pulmonaire/anatomopathologie , Hypertension artérielle pulmonaire/étiologie , Femelle , Facteur de croissance transformant bêta/métabolisme , Modèles animaux de maladie humaine , Prolifération cellulaire/génétique , Adulte d'âge moyen , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/génétique , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE