Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.896
Filtrer
1.
Allergol Immunopathol (Madr) ; 52(4): 15-20, 2024.
Article de Anglais | MEDLINE | ID: mdl-38970260

RÉSUMÉ

BACKGROUND: Pulmonary fibrosis (PF) is a chronic, progressive, and irreversible heterogeneous disease of lung interstitial tissue. To combat progression of PF, new drugs are required to be developed. Rhizoma coptidis (COP), one of the main alkaloids of Coptis chinensis, is a traditional herbal medicine used to treat various inflammatory diseases. OBJECTIVE: To investigate the possible effects of Coptisine (Cop) on the growth, inflammation, as well as FMT of TNF-ß1-induced HFL1 cells and uncover the mechanism. MATERIAL AND METHODS: Human fetal lung fibroblast 1 (HFL1) was induced using 6ng/mL TGF-ß1 as a model of pulmonary fibrosis. CCK-8, Brdu, and transwell assays indicated the effects on cell growth as well as motility. qPCR and the corresponding kits indicted the effects on cell inflammation. Immunoblot showed the effects on FMT and further confirmed the mechanism. RESULTS: Coptisine inhibits excessive growth as well as motility of TNF-ß1-induced HFL1 cells. It further inhibits inflammation and ROS levels in TNF-ß1-induced HFL1 cells. Coptisine inhibits the FMT process of TNF-ß1-induced HFL1 cells. Mechanically, coptisine promotes the Nrf2/HO-1 pathway. CONCLUSION: Coptisine can inhibit the excessive growth, inflammation as well as FMT of lung fibroblasts into myofibroblasts. It could serve as a promising drug of PF.


Sujet(s)
Berbérine , Prolifération cellulaire , Fibroblastes , Poumon , Myofibroblastes , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Berbérine/pharmacologie , Berbérine/analogues et dérivés , Myofibroblastes/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Fibroblastes/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Facteur-2 apparenté à NF-E2/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Facteur de croissance transformant bêta-1/métabolisme , Lignée cellulaire , Coptis , Heme oxygenase-1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie
2.
J Transl Med ; 22(1): 617, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961399

RÉSUMÉ

INTRODUCTION: Intrauterine adhesions (IUA) manifest as endometrial fibrosis, often causing infertility or recurrent miscarriage; however, their pathogenesis remains unclear. OBJECTIVES: This study assessed the role of Dickkopf WNT signaling pathway inhibitor 1 (DKK1) and autophagy in endometrial fibrosis, using clinical samples as well as in vitro and in vivo experiments. METHODS: Immunohistochemistry, immunofluorescence and western blot were used to determine the localization and expression of DKK1 in endometrium; DKK1 silencing and DKK1 overexpression were used to detect the biological effects of DKK1 silencing or expression in endometrial cells; DKK1 gene knockout mice were used to observe the phenotypes caused by DKK1 gene knockout. RESULTS: In patients with IUA, DKK1 and autophagy markers were down-regulated; also, α-SMA and macrophage localization were increased in the endometrium. DKK1 conditional knockout (CKO) mice showed a fibrotic phenotype with decreased autophagy and increased localization of α-SMA and macrophages in the endometrium. In vitro studies showed that DKK1 knockout (KO) suppressed the autophagic flux of endometrial stromal cells. In contrast, ectopic expression of DKK1 showed the opposite phenotype. Mechanistically, we discovered that DKK1 regulates autophagic flux through Wnt/ß-catenin and PI3K/AKT/mTOR pathways. Further studies showed that DKK1 KO promoted the secretion of interleukin (IL)-8 in exosomes, thereby promoting macrophage proliferation and metastasis. Also, in DKK1 CKO mice, treatment with autophagy activator rapamycin partially restored the endometrial fibrosis phenotype. CONCLUSION: Our findings indicated that DKK1 was a potential diagnostic marker or therapeutic target for IUA.


Sujet(s)
Autophagie , Endomètre , Exosomes , Fibrose , Protéines et peptides de signalisation intercellulaire , Macrophages , Souris knockout , Myofibroblastes , Animaux , Femelle , Protéines et peptides de signalisation intercellulaire/métabolisme , Endomètre/métabolisme , Endomètre/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Humains , Exosomes/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Souris , Souris de lignée C57BL , Adulte
3.
Int J Biol Sci ; 20(9): 3353-3371, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993568

RÉSUMÉ

Radiation-induced pulmonary fibrosis (RIPF) represents a serious complication observed in individuals undergoing thoracic radiation therapy. Currently, effective interventions for RIPF are unavailable. Prior research has demonstrated that nintedanib, a Food and Drug Administration (FDA)-approved anti-fibrotic agent for idiopathic pulmonary fibrosis, exerts therapeutic effects on chronic fibrosing interstitial lung disease. This research aimed to investigate the anti-fibrotic influences of nintedanib on RIPF and reveal the fundamental mechanisms. To assess its therapeutic impact, a mouse model of RIPF was established. The process involved nintedanib administration at various time points, both prior to and following thoracic radiation. In the RIPF mouse model, an assessment was conducted on survival rates, body weight, computed tomography features, histological parameters, and changes in gene expression. In vitro experiments were performed to discover the mechanism underlying the therapeutic impact of nintedanib on RIPF. Treatment with nintedanib, administered either two days prior or four weeks after thoracic radiation, significantly alleviated lung pathological changes, suppressed collagen deposition, and improved the overall health status of the mice. Additionally, nintedanib demonstrated significant mitigation of radiation-induced inflammatory responses in epithelial cells by inhibiting the PI3K/AKT and MAPK signaling pathways. Furthermore, nintedanib substantially inhibited fibroblast-to-myofibroblast transition by suppressing the TGF-ß/Smad and PI3K/AKT/mTOR signaling pathways. These findings suggest that nintedanib exerts preventive and therapeutic effects on RIPF by modulating multiple targets instead of a single anti-fibrotic pathway and encourage the further clinical trials to determine the efficacy of nintedanib in patients with RIPF.


Sujet(s)
Fibroblastes , Indoles , Fibrose pulmonaire , Animaux , Indoles/usage thérapeutique , Indoles/pharmacologie , Souris , Fibrose pulmonaire/étiologie , Fibrose pulmonaire/traitement médicamenteux , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Souris de lignée C57BL , Inflammation/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques
4.
Cells ; 13(13)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38994947

RÉSUMÉ

Vimentin has been reported to play diverse roles in cell processes such as spreading, migration, cell-matrix adhesion, and fibrotic transformation. Here, we assess how vimentin impacts cell spreading, morphology, and myofibroblast transformation of human corneal fibroblasts. Overall, although knockout (KO) of vimentin did not dramatically impact corneal fibroblast spreading and mechanical activity (traction force), cell elongation in response to PDGF was reduced in vimentin KO cells as compared to controls. Blocking vimentin polymerization using Withaferin had even more pronounced effects on cell spreading and also inhibited cell-induced matrix contraction. Furthermore, although absence of vimentin did not completely block TGFß-induced myofibroblast transformation, the degree of transformation and amount of αSMA protein expression was reduced. Proteomics showed that vimentin KO cells cultured in TGFß had a similar pattern of protein expression as controls. One exception included periostin, an ECM protein associated with wound healing and fibrosis in other cell types, which was highly expressed only in Vim KO cells. We also demonstrate for the first time that LRRC15, a protein previously associated with myofibroblast transformation of cancer-associated fibroblasts, is also expressed by corneal myofibroblasts. Interestingly, proteins associated with LRRC15 in other cell types, such as collagen, fibronectin, ß1 integrin and α11 integrin, were also upregulated. Overall, our data show that vimentin impacts both corneal fibroblast spreading and myofibroblast transformation. We also identified novel proteins that may regulate corneal myofibroblast transformation in the presence and/or absence of vimentin.


Sujet(s)
Cornée , Fibroblastes , Myofibroblastes , Vimentine , Humains , Vimentine/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Cornée/cytologie , Cornée/métabolisme , Facteur de croissance transformant bêta/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Withanolides/pharmacologie , Cellules cultivées
5.
Nat Commun ; 15(1): 5731, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977708

RÉSUMÉ

Neuropilin-1 (NRP1), a co-receptor for various cytokines, including TGF-ß, has been identified as a potential therapeutic target for fibrosis. However, its role and mechanism in renal fibrosis remains elusive. Here, we show that NRP1 is upregulated in distal tubular (DT) cells of patients with transplant renal insufficiency and mice with renal ischemia-reperfusion (I-R) injury. Knockout of Nrp1 reduces multiple endpoints of renal injury and fibrosis. We find that Nrp1 facilitates the binding of TNF-α to its receptor in DT cells after renal injury. This signaling results in a downregulation of lysine crotonylation of the metabolic enzyme Cox4i1, decreases cellular energetics and exacerbation of renal injury. Furthermore, by single-cell RNA-sequencing we find that Nrp1-positive DT cells secrete collagen and communicate with myofibroblasts, exacerbating acute kidney injury (AKI)-induced renal fibrosis by activating Smad3. Dual genetic deletion of Nrp1 and Tgfbr1 in DT cells better improves renal injury and fibrosis than either single knockout. Together, these results reveal that targeting of NRP1 represents a promising strategy for the treatment of AKI and subsequent chronic kidney disease.


Sujet(s)
Atteinte rénale aigüe , Fibrose , Souris knockout , Neuropiline 1 , Récepteur de type I du facteur de croissance transformant bêta , Lésion d'ischémie-reperfusion , Protéine Smad-3 , Neuropiline 1/métabolisme , Neuropiline 1/génétique , Animaux , Humains , Souris , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/génétique , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Récepteur de type I du facteur de croissance transformant bêta/génétique , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/génétique , Lésion d'ischémie-reperfusion/anatomopathologie , Protéine Smad-3/métabolisme , Protéine Smad-3/génétique , Mâle , Facteur de nécrose tumorale alpha/métabolisme , Transduction du signal , Souris de lignée C57BL , Tubules rénaux/anatomopathologie , Tubules rénaux/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Collagène/métabolisme
6.
Sci Rep ; 14(1): 15998, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987293

RÉSUMÉ

Pathological fibrosis is a significant complication of surgical procedures resulting from the accumulation of excess collagen at the site of repair which can compromise the tissue architecture and severely impede the function of the affected tissue. Few prophylactic treatments exist to counteract this process; however, the use of amniotic membrane allografts has demonstrated promising clinical outcomes. This study aimed to identify the underlying mechanism of action by utilizing relevant models that accurately represent the pathophysiology of the disease state. This study employed a pro-fibrotic in vitro system using TGFß1 stimulation and macromolecular crowding techniques to evaluate the mechanism by which amniotic membrane allografts regulate collagen biosynthesis and deposition. Following treatment with dehydrated human amnion chorion membrane (DHACM), subsequent RNA sequencing and functional enrichment with Reactome pathway analysis indicated that amniotic membranes are indeed capable of regulating genes associated with the composition and function of the extracellular matrix. Furthermore, macromolecular crowding was used in vitro to expand the evaluation to include both the effects of DHACM and a lyophilized human amnion/chorion membrane (LHACM). DHACM and LHACM regulate the TGFß pathway and myofibroblast differentiation. Additionally, both DHACM and LHACM modulate the production, secretion, and deposition of collagen type I, a primary target for pathological fibrosis. These observations support the hypothesis that amniotic membranes may interrupt pathological fibrosis by regulating collagen biosynthesis and associated pathways.


Sujet(s)
Amnios , Chorion , Collagène , Amnios/métabolisme , Humains , Chorion/métabolisme , Collagène/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Différenciation cellulaire , Matrice extracellulaire/métabolisme , Myofibroblastes/métabolisme , Fibrose , Femelle , Collagène de type I/métabolisme , Collagène de type I/génétique
7.
Cell Death Dis ; 15(7): 493, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987529

RÉSUMÉ

Lung cancer is a leading cause of cancer-related mortality globally, with a dismal 5-year survival rate, particularly for Lung Adenocarcinoma (LUAD). Mechanical changes within the tumor microenvironment, such as extracellular matrix (ECM) remodeling and fibroblast activity, play pivotal roles in cancer progression and metastasis. However, the specific impact of the basement membrane (BM) on the mechanical characteristics of LUAD remains unclear. This study aims to identify BM genes influencing internal mechanical stress in tumors, elucidating their effects on LUAD metastasis and therapy resistance, and exploring strategies to counteract these effects. Using Matrigel overlay and Transwell assays, we found that mechanical stress, mimicked by matrix application, augmented LUAD cell migration and invasion, correlating with ECM alterations and activation of the epithelial-mesenchymal transition (EMT) pathway. Employing machine learning, we developed the SVM_Score model based on relevant BM genes, which accurately predicted LUAD patient prognosis and EMT propensity across multiple datasets. Lower SVM_Scores were associated with worse survival outcomes, elevated cancer-related pathways, increased Tumor Mutation Burden, and higher internal mechanical stress in LUAD tissues. Notably, the SVM_Score was closely linked to COL5A1 expression in myofibroblasts, a key marker of mechanical stress. High COL5A1 expression from myofibroblasts promoted tumor invasiveness and EMT pathway activation in LUAD cells. Additionally, treatment with Sorafenib, which targets COL5A1 secretion, attenuated the tumor-promoting effects of myofibroblast-derived COL5A1, inhibiting LUAD cell proliferation, migration, and enhancing chemosensitivity. In conclusion, this study elucidates the complex interplay between mechanical stress, ECM alterations, and LUAD progression. The SVM_Score emerges as a robust prognostic tool reflecting tumor mechanical characteristics, while Sorafenib intervention targeting COL5A1 secretion presents a promising therapeutic strategy to mitigate LUAD aggressiveness. These findings deepen our understanding of the biomechanical aspects of LUAD and offer insights for future research and clinical applications.


Sujet(s)
Adénocarcinome pulmonaire , Collagène de type V , Transition épithélio-mésenchymateuse , Tumeurs du poumon , Myofibroblastes , Contrainte mécanique , Humains , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/traitement médicamenteux , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/anatomopathologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Collagène de type V/métabolisme , Collagène de type V/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Métastase tumorale , Souris , Microenvironnement tumoral , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Matrice extracellulaire/métabolisme
8.
Respir Res ; 25(1): 284, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026235

RÉSUMÉ

Idiopathic pulmonary fibrosis is a lethal, progressive, and irreversible condition that has become a significant focus of medical research due to its increasing incidence. This rising trend presents substantial challenges for patients, healthcare providers, and researchers. Despite the escalating burden of pulmonary fibrosis, the available therapeutic options remain limited. Currently, the United States Food and Drug Administration has approved two drugs for the treatment of pulmonary fibrosis-nintedanib and pirfenidone. However, their therapeutic effectiveness is limited, and they cannot reverse the fibrosis process. Additionally, these drugs are associated with significant side effects. Myofibroblasts play a central role in the pathophysiology of pulmonary fibrosis, significantly contributing to its progression. Consequently, strategies aimed at inhibiting myofibroblast differentiation or promoting their dedifferentiation hold promise as effective treatments. This review examines the regulation of myofibroblast dedifferentiation, exploring various signaling pathways, regulatory targets, and potential pharmaceutical interventions that could provide new directions for therapeutic development.


Sujet(s)
Dédifférenciation cellulaire , Myofibroblastes , Humains , Myofibroblastes/anatomopathologie , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Dédifférenciation cellulaire/effets des médicaments et des substances chimiques , Dédifférenciation cellulaire/physiologie , Animaux , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/traitement médicamenteux , Transduction du signal/physiologie , Antifibrotiques/usage thérapeutique , Antifibrotiques/pharmacologie , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme
9.
Zhonghua Xin Xue Guan Bing Za Zhi ; 52(7): 814-826, 2024 Jul 24.
Article de Chinois | MEDLINE | ID: mdl-39019831

RÉSUMÉ

Objective: To investigate the role and underlying mechanisms of methyltransferase (Mettl) 3 in the process of angiotensin Ⅱ (Ang Ⅱ)-induced pericyte-to-myofibroblast transdifferentiation and renal fibrosis. Methods: C57BL/6J mice were used, in cell experiments, mouse renal pericytes were isolated and cultured using magnetic bead sorting. These pericytes were then induced to transdifferentiate into myofibroblasts with 1×106 mmol/L Ang Ⅱ, which was the Ang Ⅱ group, while pericytes cultured in normal conditions served as the control group. Successful transdifferentiation was verified by immunofluorescence staining, Western blotting, and real-time reverse transcription PCR (RT-qPCR) for α-smooth muscle actin (α-SMA). The levels of m6A modifications and related enzymes (Mettl3, Mettl14), Wilms tumor 1-associated protein (WTAP), fat mass and obesity protein (FTO), ALKBH5, YTHDF1, YTHDF2, YTHDC1, YTHDC2, YTHDC3 were assessed by Dot blot, RT-qPCR and Western blot. Mettl3 expression was inhibited in cells using lentivirus-mediated Mettl3-shRNA transfection, creating sh-Mettl3 and Ang Ⅱ+sh-Mettl3 groups, while lentivirus empty vector transfection served as the negative control (Ang Ⅱ+sh-NC group). The impact of Ang Ⅱ on pericyte transdifferentiation was observed, and the expression of downstream phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway proteins, including PI3K, AKT, phosphorylated AKT at serine 473 (p-AKT (S473)), and phosphorylated AKT at threonine 308 (p-AKT (T308)), were examined. PI3K gene transcription was inhibited by co-culturing cells with actinomycin D, and the half-life of PI3K mRNA was calculated by measuring residual PI3K mRNA expression over different co-culture time. The reversibility of Mettl3 inhibition on Ang Ⅱ-induced pericyte-to-myofibroblast transdifferentiation was assessed by adding the AKT activator SC79 to the Ang Ⅱ+sh-Mettl3 group. In animal experiments, mice were divided into these groups: sham group (administered 0.9% sterile saline), Ang Ⅱ group (infused with Ang Ⅱ solution), sh-Mettl3 group (injected with Mettl3 shRNA lentivirus solution), Ang Ⅱ+sh-Mettl3 group (infused with Ang Ⅱ solution and injected with Mettl3 shRNA lentivirus solution), and Ang Ⅱ+sh-Mettl3+SC79 group (administered Ang Ⅱ solution and Mettl3 shRNA lentivirus, with an additional injection of SC79). Each group consisted of six subject mice. Blood pressure was measured using the tail-cuff method before and after surgery, and serum creatinine, urea, and urinary albumin levels were determined 4 weeks post-surgery. Kidney tissues were collected at 28 days and stained using hematoxylin-eosin (HE) and Masson's trichrome to assess the extent of renal fibrosis. Results: Primary renal pericytes were successfully obtained by magnetic bead sorting, and intervened with 1×106 mmol/L Ang Ⅱ for 48 hours to induce pericyte-to-myofibroblast transdifferentiation. Dot blot results indicated higher m6A modification levels in the Ang Ⅱ group compared to the control group (P<0.05). RT-qPCR and Western blot results showed upregulation of Mettl3 mRNA and protein levels in the Ang Ⅱ group compared to the control group (both P<0.05). In the Ang Ⅱ+sh-Mettl3 group, Mettl3 protein expression was lower than that in the Ang Ⅱ group, with reduced expression levels of α-SMA, vimentin, desmin, fibroblast agonist protein (FAPa) and type Ⅰ collagen (all P<0.05). Compared to the control group, PI3K mRNA expression level was elevated in the Ang Ⅱ group, along with increased p-AKT (S473) and p-AKT (T308) expressions. In the Ang Ⅱ+sh-Mettl3 group, PI3K mRNA expression and p-AKT (S473) and p-AKT (T308) levels were decreased (all P<0.05). The half-life of PI3K mRNA was shorter in the Ang Ⅱ+sh-Mettl3 group than that in the Ang Ⅱ+sh-NC group (2.34 h vs. 3.42 h). The ameliorative effect of Mettl3 inhibition on Ang Ⅱ-induced pericyte-to-myofibroblast transdifferentiation was reversible by SC79. Animal experiments showed higher blood pressure, serum creatinine, urea, and 24-hour urinary protein levels, and a larger fibrosis area in the Ang Ⅱ group compared to the sham group (all P<0.05). The fibrosis area was smaller in the Ang Ⅱ+sh-Mettl3 group than that in the Ang Ⅱ group (P<0.05), but increased again upon addition of SC79. Conclusion: Mettl3-mediated RNA m6A epigenetic regulation is involved in Ang Ⅱ-induced pericyte-to-myofibroblast transdifferentiation and renal fibrosis, potentially by affecting PI3K stability and regulating the PI3K/AKT signaling pathway.


Sujet(s)
Angiotensine-II , Transdifférenciation cellulaire , Methyltransferases , Souris de lignée C57BL , Myofibroblastes , Péricytes , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Péricytes/métabolisme , Methyltransferases/métabolisme , Souris , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Angiotensine-II/pharmacologie , Myofibroblastes/métabolisme , Rein , Cellules cultivées
10.
Mol Biol Cell ; 35(8): ar114, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38985514

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF), one of the most common forms of interstitial lung disease, is a poorly understood, chronic, and often fatal fibroproliferative condition with only two FDA-approved medications. Understanding the pathobiology of the fibroblast in IPF is critical to evaluating and discovering novel therapeutics. Using a decellularized lung matrix derived from patients with IPF, we generate three-dimensional hydrogels as in vitro models of lung physiology and characterize the phenotype of fibroblasts seeded into the hydrogels. When cultured in IPF extracellular matrix hydrogels, IPF fibroblasts display differential contractility compared with their normal counterparts, lose the classical myofibroblast marker α-smooth muscle actin, and increase expression of proinflammatory cytokines compared with fibroblasts seeded two-dimensionally on tissue culture dishes. We validate this proinflammatory state in fibroblast-conditioned media studies with monocytes and monocyte-derived macrophages. These findings add to a growing understanding of the lung microenvironment effect on fibroblast phenotypes, shed light on the potential role of fibroblasts as immune signaling hubs during lung fibrosis, and suggest intervention in fibroblast-immune cell cross-talk as a possible novel therapeutic avenue.


Sujet(s)
Matrice extracellulaire , Fibroblastes , Hydrogels , Fibrose pulmonaire idiopathique , Poumon , Humains , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibroblastes/métabolisme , Poumon/anatomopathologie , Poumon/métabolisme , Matrice extracellulaire/métabolisme , Cytokines/métabolisme , Macrophages/métabolisme , Myofibroblastes/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Cellules cultivées , Milieux de culture conditionnés/pharmacologie , Actines/métabolisme , Monocytes/métabolisme
11.
FASEB J ; 38(13): e23749, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38953707

RÉSUMÉ

Pulmonary fibrosis is a formidable challenge in chronic and age-related lung diseases. Myofibroblasts secrete large amounts of extracellular matrix and induce pro-repair responses during normal wound healing. Successful tissue repair results in termination of myofibroblast activity via apoptosis; however, some myofibroblasts exhibit a senescent phenotype and escape apoptosis, causing over-repair that is characterized by pathological fibrotic scarring. Therefore, the removal of senescent myofibroblasts using senolytics is an important method for the treatment of pulmonary fibrosis. Procyanidin C1 (PCC1) has recently been discovered as a senolytic compound with very low toxicity and few side effects. This study aimed to determine whether PCC1 could improve lung fibrosis by promoting apoptosis in senescent myofibroblasts and to investigate the mechanisms involved. The results showed that PCC1 attenuates bleomycin (BLM)-induced pulmonary fibrosis in mice. In addition, we found that PCC1 inhibited extracellular matrix deposition and promoted the apoptosis of senescent myofibroblasts by increasing PUMA expression and activating the BAX signaling pathway. Our findings represent a new method of pulmonary fibrosis management and emphasize the potential of PCC1 as a senotherapeutic agent for the treatment of pulmonary fibrosis, providing hope for patients with pulmonary fibrosis worldwide. Our results advance our understanding of age-related diseases and highlight the importance of addressing cellular senescence in treatment.


Sujet(s)
Bléomycine , Catéchine , Vieillissement de la cellule , Souris de lignée C57BL , Myofibroblastes , Fibrose pulmonaire , Animaux , Bléomycine/toxicité , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Souris , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Catéchine/pharmacologie , Catéchine/analogues et dérivés , Proanthocyanidines/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Mâle , Biflavonoïdes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
12.
Nat Commun ; 15(1): 5895, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003267

RÉSUMÉ

Autoimmune thyroid diseases (AITD) such as Graves' disease (GD) or Hashimoto's thyroiditis (HT) are organ-specific diseases that involve complex interactions between distinct components of thyroid tissue. Here, we use spatial transcriptomics to explore the molecular architecture, heterogeneity and location of different cells present in the thyroid tissue, including thyroid follicular cells (TFCs), stromal cells such as fibroblasts, endothelial cells, and thyroid infiltrating lymphocytes. We identify damaged antigen-presenting TFCs with upregulated CD74 and MIF expression in thyroid samples from AITD patients. Furthermore, we discern two main fibroblast subpopulations in the connective tissue including ADIRF+ myofibroblasts, mainly enriched in GD, and inflammatory fibroblasts, enriched in HT patients. We also demonstrate an increase of fenestrated PLVAP+ vessels in AITD, especially in GD. Our data unveil stromal and thyroid epithelial cell subpopulations that could play a role in the pathogenesis of AITD.


Sujet(s)
Antigènes de différenciation des lymphocytes B , Maladie de Basedow , Maladie de Hashimoto , Glande thyroide , Humains , Maladie de Basedow/anatomopathologie , Maladie de Basedow/immunologie , Maladie de Basedow/génétique , Maladie de Basedow/métabolisme , Glande thyroide/anatomopathologie , Glande thyroide/métabolisme , Maladie de Hashimoto/anatomopathologie , Maladie de Hashimoto/immunologie , Maladie de Hashimoto/métabolisme , Maladie de Hashimoto/génétique , Antigènes de différenciation des lymphocytes B/métabolisme , Antigènes de différenciation des lymphocytes B/génétique , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Antigènes d'histocompatibilité de classe II/métabolisme , Antigènes d'histocompatibilité de classe II/génétique , Cellules épithéliales thyroïdiennes/métabolisme , Cellules épithéliales thyroïdiennes/anatomopathologie , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Transcriptome , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Femelle , Facteurs inhibiteurs de la migration des macrophages , Intramolecular oxidoreductases
13.
Theranostics ; 14(9): 3603-3622, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948058

RÉSUMÉ

Background: Myofibroblasts (MYFs) are generally considered the principal culprits in excessive extracellular matrix deposition and scar formation in the pathogenesis of lung fibrosis. Lipofibroblasts (LIFs), on the other hand, are defined by their lipid-storing capacity and are predominantly found in the alveolar regions of the lung. They have been proposed to play a protective role in lung fibrosis. We previously reported that a LIF to MYF reversible differentiation switch occurred during fibrosis formation and resolution. In this study, we tested whether WI-38 cells, a human embryonic lung fibroblast cell line, could be used to study fibroblast differentiation towards the LIF or MYF phenotype and whether this could be relevant for idiopathic pulmonary fibrosis (IPF). Methods: Using WI-38 cells, Fibroblast (FIB) to MYF differentiation was triggered using TGF-ß1 treatment and FIB to LIF differentiation using Metformin treatment. We also analyzed the MYF to LIF and LIF to MYF differentiation by pre-treating the WI-38 cells with TGF-ß1 or Metformin respectively. We used IF, qPCR and bulk RNA-Seq to analyze the phenotypic and transcriptomic changes in the cells. We correlated our in vitro transcriptome data from WI-38 cells (obtained via bulk RNA sequencing) with the transcriptomic signature of LIFs and MYFs derived from the IPF cell atlas as well as with our own single-cell transcriptomic data from IPF patients-derived lung fibroblasts (LF-IPF) cultured in vitro. We also carried out alveolosphere assays to evaluate the ability of the proposed LIF and MYF cells to support the growth of alveolar epithelial type 2 cells. Results: WI-38 cells and LF-IPF display similar phenotypical and gene expression responses to TGF-ß1 and Metformin treatment. Bulk RNA-Seq analysis of WI-38 cells and LF-IPF treated with TGF-ß1, or Metformin indicate similar transcriptomic changes. We also show the partial conservation of the LIF and MYF signature extracted from the Habermann et al. scRNA-seq dataset in WI-38 cells treated with Metformin or TGF-ß1, respectively. Alveolosphere assays indicate that LIFs enhance organoid growth, while MYFs inhibit organoid growth. Finally, we provide evidence supporting the MYF to LIF and LIF to MYF reversible switch using WI-38 cells. Conclusions: WI-38 cells represent a versatile and reliable model to study the intricate dynamics of fibroblast differentiation towards the MYF or LIF phenotype associated with lung fibrosis formation and resolution, providing valuable insights to drive future research.


Sujet(s)
Différenciation cellulaire , Fibroblastes , Fibrose pulmonaire idiopathique , Myofibroblastes , Facteur de croissance transformant bêta-1 , Humains , Myofibroblastes/métabolisme , Fibroblastes/métabolisme , Lignée cellulaire , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Poumon/anatomopathologie , Poumon/cytologie , Transcriptome , Metformine/pharmacologie , Plasticité cellulaire/effets des médicaments et des substances chimiques , Phénotype
14.
Cells ; 13(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38920695

RÉSUMÉ

Aberrant sialylation with overexpression of the homopolymeric glycan polysialic acid (polySia) was recently reported in fibroblasts from fibrotic skin lesions. Yet, whether such a rise in polySia levels or sialylation in general may be functionally implicated in profibrotic activation of fibroblasts and their transition to myofibroblasts remains unknown. Therefore, we herein explored whether inhibition of sialylation could interfere with the process of skin fibroblast-to-myofibroblast transition induced by the master profibrotic mediator transforming growth factor ß1 (TGFß1). Adult human skin fibroblasts were pretreated with the competitive pan-sialyltransferase inhibitor 3-Fax-peracetyl-Neu5Ac (3-Fax) before stimulation with recombinant human TGFß1, and then analyzed for polySia expression, cell viability, proliferation, migratory ability, and acquisition of myofibroblast-like morphofunctional features. Skin fibroblast stimulation with TGFß1 resulted in overexpression of polySia, which was effectively blunted by 3-Fax pre-administration. Pretreatment with 3-Fax efficiently lessened TGFß1-induced skin fibroblast proliferation, migration, changes in cell morphology, and phenotypic and functional differentiation into myofibroblasts, as testified by a significant reduction in FAP, ACTA2, COL1A1, COL1A2, and FN1 gene expression, and α-smooth muscle actin, N-cadherin, COL1A1, and FN-EDA protein levels, as well as a reduced contractile capability. Moreover, skin fibroblasts pre-administered with 3-Fax displayed a significant decrease in Smad3-dependent canonical TGFß1 signaling. Collectively, our in vitro findings demonstrate for the first time that aberrant sialylation with increased polySia levels has a functional role in skin fibroblast-to-myofibroblast transition and suggest that competitive sialyltransferase inhibition might offer new therapeutic opportunities against skin fibrosis.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Fibroblastes , Myofibroblastes , Acides sialiques , Peau , Facteur de croissance transformant bêta-1 , Humains , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/pharmacologie , Peau/métabolisme , Peau/anatomopathologie , Acides sialiques/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Sialyltransferases/métabolisme , Sialyltransferases/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Cellules cultivées
15.
Int Immunopharmacol ; 136: 112338, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38850787

RÉSUMÉ

Cardiac fibrosis is a typical feature of cardiac pathological remodeling, which is associated with adverse clinical outcomes and has no effective therapy. Nicotine is an important risk factor for cardiac fibrosis, yet its underlying molecular mechanism remains poorly understood. This study aimed to identify its potential molecular mechanism in nicotine-induced cardiac fibrosis. Our results showed nicotine exposure led to the proliferation and transformation of cardiac fibroblasts (CFs) into myofibroblasts (MFs) by impairing autophagy flux. Through the use of drug affinity responsive target stability (DARTS) assay, cellular thermal shift assay (CETSA), and surface plasmon resonance (SPR) technology, it was discovered that nicotine directly increased the stability and protein levels of lactate dehydrogenase A (LDHA) by binding to it. Nicotine treatment impaired autophagy flux by regulating the AMPK/mTOR signaling pathway, impeding the nuclear translocation of transcription factor EB (TFEB), and reducing the activity of cathepsin B (CTSB). In vivo, nicotine treatment exacerbated cardiac fibrosis induced in spontaneously hypertensive rats (SHR) and worsened cardiac function. Interestingly, the absence of LDHA reversed these effects both in vitro and in vivo. Our study identified LDHA as a novel nicotine-binding protein that plays a crucial role in mediating cardiac fibrosis by blocking autophagy flux. The findings suggest that LDHA could potentially serve as a promising target for the treatment of cardiac fibrosis.


Sujet(s)
Autophagie , Fibrose , Nicotine , Animaux , Autophagie/effets des médicaments et des substances chimiques , Rats , Mâle , Rats de lignée SHR , Transduction du signal/effets des médicaments et des substances chimiques , Myocarde/anatomopathologie , Myocarde/métabolisme , Lactate dehydrogenase 5/métabolisme , Cellules cultivées , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Rat Sprague-Dawley
16.
Sci Adv ; 10(25): eadk8501, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38905342

RÉSUMÉ

Single-cell technology has allowed researchers to probe tissue complexity and dynamics at unprecedented depth in health and disease. However, the generation of high-dimensionality single-cell atlases and virtual three-dimensional tissues requires integrated reference maps that harmonize disparate experimental designs, analytical pipelines, and taxonomies. Here, we present a comprehensive single-cell transcriptome integration map of cardiac fibrosis, which underpins pathophysiology in most cardiovascular diseases. Our findings reveal similarity between cardiac fibroblast (CF) identities and dynamics in ischemic versus pressure overload models of cardiomyopathy. We also describe timelines for commitment of activated CFs to proliferation and myofibrogenesis, profibrotic and antifibrotic polarization of myofibroblasts and matrifibrocytes, and CF conservation across mouse and human healthy and diseased hearts. These insights have the potential to inform knowledge-based therapies.


Sujet(s)
Fibroblastes , Fibrose , Analyse sur cellule unique , Transcriptome , Animaux , Analyse sur cellule unique/méthodes , Humains , Fibroblastes/métabolisme , Souris , Myocarde/métabolisme , Myocarde/anatomopathologie , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Analyse de profil d'expression de gènes
17.
Commun Biol ; 7(1): 736, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890483

RÉSUMÉ

Organ fibrosis causes collagen fiber overgrowth and impairs organ function. Cardiac fibrosis after myocardial infarction impairs cardiac function significantly, pulmonary fibrosis reduces gas exchange efficiency, and liver fibrosis disturbs the natural function of the liver. Its development is associated with the differentiation of fibroblasts into myofibroblasts and increased collagen synthesis. Fibrosis has organ specificity, defined by the heterogeneity of fibroblasts. Although this heterogeneity is established during embryonic development, it has not been defined yet. Fibroblastic differentiation of induced pluripotent stem cells (iPSCs) recapitulates the process by which fibroblasts acquire diversity. Here, we differentiated iPSCs into cardiac, hepatic, and dermal fibroblasts and analyzed their properties using single-cell RNA sequencing. We observed characteristic subpopulations with different ratios in each organ-type fibroblast group, which contained both resting and distinct ACTA2+ myofibroblasts. These findings provide crucial information on the ontogeny-based heterogeneity of fibroblasts, leading to the development of therapeutic strategies to control fibrosis.


Sujet(s)
Différenciation cellulaire , Fibroblastes , Cellules souches pluripotentes induites , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Fibroblastes/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/cytologie , Cellules cultivées , Analyse sur cellule unique , Fibrose
18.
Circ Res ; 135(2): 280-297, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38847080

RÉSUMÉ

BACKGROUND: Heart failure (HF) is one of the leading causes of mortality worldwide. Extracellular vesicles, including small extracellular vesicles or exosomes, and their molecular cargo are known to modulate cell-to-cell communication during multiple cardiac diseases. However, the role of systemic extracellular vesicle biogenesis inhibition in HF models is not well documented and remains unclear. METHODS: We investigated the role of circulating exosomes during cardiac dysfunction and remodeling in a mouse transverse aortic constriction (TAC) model of HF. Importantly, we investigate the efficacy of tipifarnib, a recently identified exosome biogenesis inhibitor that targets the critical proteins (Rab27a [Ras associated binding protein 27a], nSMase2 [neutral sphingomyelinase 2], and Alix [ALG-2-interacting protein X]) involved in exosome biogenesis for this mouse model of HF. In this study, 10-week-old male mice underwent TAC surgery were randomly assigned to groups with and without tipifarnib treatment (10 mg/kg 3 times/wk) and monitored for 8 weeks, and a comprehensive assessment was conducted through performed echocardiographic, histological, and biochemical studies. RESULTS: TAC significantly elevated circulating plasma exosomes and markedly increased cardiac left ventricular dysfunction, cardiac hypertrophy, and fibrosis. Furthermore, injection of plasma exosomes from TAC mice induced left ventricular dysfunction and cardiomyocyte hypertrophy in uninjured mice without TAC. On the contrary, treatment of tipifarnib in TAC mice reduced circulating exosomes to baseline and remarkably improved left ventricular functions, hypertrophy, and fibrosis. Tipifarnib treatment also drastically altered the miRNA profile of circulating post-TAC exosomes, including miR 331-5p, which was highly downregulated both in TAC circulating exosomes and in TAC cardiac tissue. Mechanistically, miR 331-5p is crucial for inhibiting the fibroblast-to-myofibroblast transition by targeting HOXC8, a critical regulator of fibrosis. Tipifarnib treatment in TAC mice upregulated the expression of miR 331-5p that acts as a potent repressor for one of the fibrotic mechanisms mediated by HOXC8. CONCLUSIONS: Our study underscores the pathological role of exosomes in HF and fibrosis in response to pressure overload. Tipifarnib-mediated inhibition of exosome biogenesis and cargo sorting may serve as a viable strategy to prevent progressive cardiac remodeling in HF.


Sujet(s)
Vésicules extracellulaires , Défaillance cardiaque , Quinolinone , Animaux , Mâle , Souris , Cardiotoniques/pharmacologie , Cardiotoniques/usage thérapeutique , Modèles animaux de maladie humaine , Vésicules extracellulaires/effets des médicaments et des substances chimiques , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/prévention et contrôle , Quinolinone/pharmacologie , Quinolinone/usage thérapeutique , Répartition aléatoire , Régulation positive/effets des médicaments et des substances chimiques , microARN , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme
19.
Cell Metab ; 36(6): 1287-1301.e7, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38838641

RÉSUMÉ

Adipocytes in dermis are considered to be important participants in skin repair and regeneration, but the role of subcutaneous white adipose tissue (sWAT) in skin repair is poorly understood. Here, we revealed the dynamic changes of sWAT during wound healing process. Lineage-tracing mouse studies revealed that sWAT would enter into the large wound bed and participate in the formation of granulation tissue. Moreover, sWAT undergoes beiging after skin injury. Inhibition of sWAT beiging by genetically silencing PRDM16, a key regulator to beiging, hindered wound healing process. The transcriptomics results suggested that beige adipocytes in sWAT abundantly express neuregulin 4 (NRG4), which regulated macrophage polarization and the function of myofibroblasts. In diabetic wounds, the beiging of sWAT was significantly suppressed. Thus, adipocytes from sWAT regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes.


Sujet(s)
Tissu adipeux blanc , Peau , Cicatrisation de plaie , Animaux , Tissu adipeux blanc/métabolisme , Souris , Peau/métabolisme , Peau/anatomopathologie , Souris de lignée C57BL , Graisse sous-cutanée/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Neurégulines/métabolisme , Neurégulines/génétique , Mâle , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Tissu adipeux brun/métabolisme , Adipocytes beiges/métabolisme , Macrophages/métabolisme , Humains , Myofibroblastes/métabolisme
20.
Cell Death Dis ; 15(6): 391, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830870

RÉSUMÉ

Tissue injury causes activation of mesenchymal lineage cells into wound-repairing myofibroblasts (MFs), whose uncontrolled activity ultimately leads to fibrosis. Although this process is triggered by deep metabolic and transcriptional reprogramming, functional links between these two key events are not yet understood. Here, we report that the metabolic sensor post-translational modification O-linked ß-D-N-acetylglucosaminylation (O-GlcNAcylation) is increased and required for myofibroblastic activation. Inhibition of protein O-GlcNAcylation impairs archetypal myofibloblast cellular activities including extracellular matrix gene expression and collagen secretion/deposition as defined in vitro and using ex vivo and in vivo murine liver injury models. Mechanistically, a multi-omics approach combining proteomic, epigenomic, and transcriptomic data mining revealed that O-GlcNAcylation controls the MF transcriptional program by targeting the transcription factors Basonuclin 2 (BNC2) and TEA domain transcription factor 4 (TEAD4) together with the Yes-associated protein 1 (YAP1) co-activator. Indeed, inhibition of protein O-GlcNAcylation impedes their stability leading to decreased functionality of the BNC2/TEAD4/YAP1 complex towards promoting activation of the MF transcriptional regulatory landscape. We found that this involves O-GlcNAcylation of BNC2 at Thr455 and Ser490 and of TEAD4 at Ser69 and Ser99. Altogether, this study unravels protein O-GlcNAcylation as a key determinant of myofibroblastic activation and identifies its inhibition as an avenue to intervene with fibrogenic processes.


Sujet(s)
Myofibroblastes , Transduction du signal , Myofibroblastes/métabolisme , Animaux , Souris , Humains , Fibrose/métabolisme , Facteurs de transcription/métabolisme , Protéines de signalisation YAP/métabolisme , Souris de lignée C57BL , Facteurs de transcription à domaine TEA/métabolisme , Mâle , Maturation post-traductionnelle des protéines , Acétyl-glucosamine/métabolisme , Transcription génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE