Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.796
Filtrer
1.
Sci Adv ; 10(32): eadl5473, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39121212

RÉSUMÉ

Despite advancements in antifibrotic therapy, idiopathic pulmonary fibrosis (IPF) remains a medical condition with unmet needs. Single-cell RNA sequencing (scRNA-seq) has enhanced our understanding of IPF but lacks the cellular tissue context and gene expression localization that spatial transcriptomics provides. To bridge this gap, we profiled IPF and control patient lung tissue using spatial transcriptomics, integrating the data with an IPF scRNA-seq atlas. We identified three disease-associated niches with unique cellular compositions and localizations. These include a fibrotic niche, consisting of myofibroblasts and aberrant basaloid cells, located around airways and adjacent to an airway macrophage niche in the lumen, containing SPP1+ macrophages. In addition, we identified an immune niche, characterized by distinct lymphoid cell foci in fibrotic tissue, surrounded by remodeled endothelial vessels. This spatial characterization of IPF niches will facilitate the identification of drug targets that disrupt disease-driving niches and aid in the development of disease relevant in vitro models.


Sujet(s)
Fibrose pulmonaire idiopathique , Poumon , Transcriptome , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/génétique , Humains , Poumon/anatomopathologie , Poumon/métabolisme , Macrophages/métabolisme , Analyse sur cellule unique , Analyse de profil d'expression de gènes , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie
2.
Transl Vis Sci Technol ; 13(8): 22, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39133495

RÉSUMÉ

Purpose: The purpose of this study was to evaluate the safety and efficacy of topical losartan in the therapeutic treatment of established corneal scaring fibrosis at 1 month after alkali burn in rabbits. Methods: Standardized alkali burns were performed in 1 eye of 24 rabbits with 0.75N NaOH for 15 seconds. Corneas were allowed to heal and develop scaring of the cornea for 1 month. Twelve eyes per group were treated with 50 µL of topical 0.8 mg/mL losartan in balanced salt solution (BSS), pH 7.0, and 12 eyes were treated with vehicle BSS 6 times per day. Six corneas were analyzed at 1 week or 1 month in each group. Standardized slit lamp photographs were obtained at the end point for each cornea and opacity was quantitated using ImageJ. Corneoscleral rims were cryofixed in optimum cutting temperature (OCT) solution and combined duplex immunohistochemistry for myofibroblast marker alpha-smooth muscle actin (α-SMA), mesenchymal cell marker vimentin, and TUNEL assay for apoptosis was performed on all corneas. Results: Topical losartan was effective in the treatment of established stromal fibrosis following alkali burn injury to the rabbit cornea. Stromal myofibroblast density was decreased and stromal cell apoptosis was increased (included both α-SMA-positive myofibroblasts and α-SMA-negative, vimentin-positive cells) at both 1 week and 1 month in the topical losartan-treated compared with vehicle-treated groups. Conclusions: Topical losartan is effective in the treatment of established stromal fibrosis in rabbits. Most myofibroblasts disappear from the stroma within the first month of losartan treatment. Longer treatment with topical losartan is needed to allow time for corneal fibroblast regeneration of the epithelial basement membrane (in coordination with epithelial cells) and the removal of disordered extracellular matrix produced by myofibroblasts.


Sujet(s)
Brûlures chimiques , Brûlures oculaires , Fibrose , Losartan , Animaux , Lapins , Losartan/pharmacologie , Losartan/administration et posologie , Losartan/usage thérapeutique , Fibrose/traitement médicamenteux , Brûlures chimiques/traitement médicamenteux , Brûlures chimiques/anatomopathologie , Brûlures oculaires/traitement médicamenteux , Brûlures oculaires/anatomopathologie , Brûlures oculaires/induit chimiquement , Modèles animaux de maladie humaine , Apoptose/effets des médicaments et des substances chimiques , Antagonistes du récepteur de type 1 de l'angiotensine-II/administration et posologie , Antagonistes du récepteur de type 1 de l'angiotensine-II/pharmacologie , Antagonistes du récepteur de type 1 de l'angiotensine-II/usage thérapeutique , Hydroxyde de sodium , Maladies de la cornée/traitement médicamenteux , Maladies de la cornée/anatomopathologie , Solutions ophtalmiques/usage thérapeutique , Solutions ophtalmiques/administration et posologie , Cornée/effets des médicaments et des substances chimiques , Cornée/anatomopathologie , Méthode TUNEL , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/anatomopathologie , Actines/métabolisme , Mâle , Stroma de la cornée/effets des médicaments et des substances chimiques , Stroma de la cornée/anatomopathologie , Stroma de la cornée/métabolisme , Administration par voie topique , Vimentine/métabolisme , Cicatrisation de plaie/effets des médicaments et des substances chimiques
3.
Sci Rep ; 14(1): 18840, 2024 08 13.
Article de Anglais | MEDLINE | ID: mdl-39138336

RÉSUMÉ

The combination of lineage tracing and immunohistochemistry has helped to identify subpopulations and fate of hepatic stellate cells (HSC) in murine liver. HSC are sinusoidal pericytes that act as myofibroblast precursors after liver injury. Single cell RNA sequencing approaches have recently helped to differentiate central and portal HSC. A specific Cre line to lineage trace portal HSC has not yet been described. We used three Cre lines (Lrat-Cre, PDGFRß-CreERT2 and SMMHC-CreERT2) known to label mesenchymal cells including HSC in combination with a tdTomato-expressing reporter. All three Cre lines labeled populations of HSC as well as smooth muscle cells (SMC). Using the SMMHC-CreERT2, we identified a subtype of HSC in the periportal area of the hepatic lobule (termed zone 1-HSC). We lineage traced tdTomato-expressing zone 1-HSC over 1 year, described fibrotic behavior in two fibrosis models and investigated their possible role during fibrosis. This HSC subtype resides in zone 1 under healthy conditions; however, zonation is disrupted in preclinical models of liver fibrosis (CCl4 and MASH). Zone 1-HSC do not transform into αSMA-expressing myofibroblasts. Rather, they participate in sinusoidal capillarization. We describe a novel subtype of HSC restricted to zone 1 under physiological conditions and its possible function after liver injury. In contrast to the accepted notion, this HSC subtype does not transform into αSMA-positive myofibroblasts; rather, zone 1-HSC adopt properties of capillary pericytes, thereby participating in sinusoidal capillarization.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Myofibroblastes , Animaux , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Souris , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Foie/anatomopathologie , Foie/métabolisme , Péricytes/métabolisme , Péricytes/anatomopathologie , Lignage cellulaire , Mâle , Différenciation cellulaire , Modèles animaux de maladie humaine , Souris de lignée C57BL
4.
Biomed Pharmacother ; 178: 117246, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39096617

RÉSUMÉ

Idiopathic pulmonary fibrosis is a progressive lung disease characterized by excessive extracellular matrix accumulation and myofibroblast proliferation with limited treatment options available. M2 macrophages are pivotal in pulmonary fibrosis, where they induce the epithelial-to-mesenchymal and fibroblast-to-myofibroblast transitions. In this study, we evaluated whether MEL-dKLA, a hybrid peptide that can eliminate M2 macrophages, could attenuate pulmonary fibrosis in a cell co-culture system and in a bleomycin-induced mouse model. Our findings demonstrated that the removal of M2 macrophages using MEL-dKLA stimulated reprogramming to an antifibrotic environment, which effectively suppressed epithelial-to-mesenchymal and fibroblast-to-myofibroblast transition responses in lung epithelial and fibroblast cells and reduced extracellular matrix accumulation both in vivo and in vitro. Moreover, MEL-dKLA exhibited antifibrotic efficacy without damaging tissue-resident macrophages in the bleomycin-induced mouse model. Collectively, our findings suggest that MEL-dKLA may be a new therapeutic option for the treatment of idiopathic pulmonary fibrosis.


Sujet(s)
Bléomycine , Fibrose pulmonaire idiopathique , Macrophages , Souris de lignée C57BL , Animaux , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/induit chimiquement , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Modèles animaux de maladie humaine , Mâle , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/traitement médicamenteux , Techniques de coculture , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Fibroblastes/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Matrice extracellulaire/métabolisme , Myofibroblastes/anatomopathologie , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Cellules RAW 264.7
5.
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
6.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39063116

RÉSUMÉ

The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by excessive deposition of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs). In the healthy liver, quiescent HSCs metabolize and store retinoids. Upon fibrogenic activation, quiescent HSCs transdifferentiate into myofibroblasts; lose their vitamin A; upregulate α-smooth muscle actin; and produce proinflammatory soluble mediators, collagens, and inhibitors of ECM degradation. Activated HSCs are the main effector cells during hepatic fibrogenesis. In addition, the accumulation and activation of profibrogenic macrophages in response to hepatocyte death play a critical role in the initiation of HSC activation and survival. The main source of myofibroblasts is resident HSCs. Activated HSCs migrate to the site of active fibrogenesis to initiate the formation of a fibrous scar. Single-cell technologies revealed that quiescent HSCs are highly homogenous, while activated HSCs/myofibroblasts are much more heterogeneous. The complex process of inflammation results from the response of various hepatic cells to hepatocellular death and inflammatory signals related to intrahepatic injury pathways or extrahepatic mediators. Inflammatory processes modulate fibrogenesis by activating HSCs and, in turn, drive immune mechanisms via cytokines and chemokines. Increasing evidence also suggests that cellular stress responses contribute to fibrogenesis. Recent data demonstrated that LF can revert even at advanced stages of cirrhosis if the underlying cause is eliminated, which inhibits the inflammatory and profibrogenic cells. However, despite numerous clinical studies on plausible drug candidates, an approved antifibrotic therapy still remains elusive. This state-of-the-art review presents cellular and molecular mechanisms involved in hepatic fibrogenesis and its resolution, as well as comprehensively discusses the drivers linking liver injury to chronic liver inflammation and LF.


Sujet(s)
Cellules étoilées du foie , Cirrhose du foie , Humains , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Animaux , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie
7.
ACS Appl Mater Interfaces ; 16(29): 37530-37544, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-38989714

RÉSUMÉ

Contrary to the initial belief that myofibroblasts are terminally differentiated cells, myofibroblasts have now been widely recognized as an activation state that is reversible. Therefore, strategies targeting myofibroblast to be a quiescent state may be an effective way for antihypertrophic scar therapy. Graphene quantum dots (GQDs), a novel zero-dimensional and carbon-based nanomaterial, have recently garnered significant interest in nanobiomedicine, owing to their excellent biocompatibility, tunable photoluminescence, and superior physiological stability. Although multiple nanoparticles have been used to alleviate hypertrophic scars, a GQD-based therapy has not been reported. Our in vivo studies showed that GQDs exhibited significant antiscar efficacy, with scar appearance improvement, collagen reduction and rearrangement, and inhibition of myofibroblast overproliferation. Further in vitro experiments revealed that GQDs inhibited α-SMA expression, collagen synthesis, and cell proliferation and migration, inducing myofibroblasts to become quiescent fibroblasts. Mechanistic studies have demonstrated that the effect of GQDs on myofibroblast proliferation blocked cell cycle progression by disrupting the cyclin-CDK-E2F axis. This study suggests that GQDs, which promote myofibroblast-to-fibroblast transition, could be a novel antiscar nanomedicine for the treatment of hypertrophic scars and other types of pathological fibrosis.


Sujet(s)
Prolifération cellulaire , Cicatrice hypertrophique , Graphite , Myofibroblastes , Boîtes quantiques , Boîtes quantiques/composition chimique , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/anatomopathologie , Myofibroblastes/métabolisme , Graphite/composition chimique , Graphite/pharmacologie , Cicatrice hypertrophique/traitement médicamenteux , Cicatrice hypertrophique/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Animaux , Humains , Souris , Collagène/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques
8.
Ann Clin Lab Sci ; 54(3): 363-370, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-39048163

RÉSUMÉ

OBJECTIVE: During the progression of chronic idiopathic pulmonary fibrosis (IPF), maladaptive tissue remodeling including excessive extracellular matrix (ECM) deposition occurs, which eventually leads to architectural distortion and loss of organ function in organ fibrosis. ADAM15, which is highly expressed in the developing lungs and kidneys, is a transmembrane-anchored multidomain protein belonging to the family of metalloproteinases. Compared to the extensive studies about functions of matrix metalloproteinases (MMPs), less are discussed about ADAM15, particularly in function and mechanism involving fibrogenesis. Our study aims to fill in this gap. METHODS: We identified ADAM15 as a novel antifibrotic mediator in lung fibrosis. We found that ADAM15 has cross-talks with transforming growth factor-ß1 (TGF-ß1), which is the most potent profibrotic mediator. We provided molecular and translational evidence that knockdown of ADAM15 accelerated fibrogenic response induced by TGF-ß1 and upregulation of ADAM15 rescued TGF-ß1-induced myofibroblast activation in part. RESULTS: Overexpression of ADAM15 ameliorates fibrotic changes and ADAM15 deficiency exacerbates changes from fibroblast to myofibroblast in NIH/3T3. Results were also presented and identified by the intuitive immunofluorescence staining. CONCLUSION: In this study, we uncover a new molecular mechanism of tissue fibrogenesis and identify ADAM15 as a potential therapeutic target in the treatment of fibrotic diseases.


Sujet(s)
Protéines ADAM , Matrice extracellulaire , Fibroblastes , Protéines membranaires , Facteur de croissance transformant bêta-1 , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Souris , Fibroblastes/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines ADAM/métabolisme , Protéines ADAM/génétique , Matrice extracellulaire/métabolisme , Humains , Cellules NIH 3T3 , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme
9.
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
10.
Int Immunopharmacol ; 139: 112705, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39029235

RÉSUMÉ

Fibrosis is not a disease but rather an outcome of the pathological tissue repair response. Many myofibroblasts are activated which lead to the excessive accumulation of extracellular matrix components such as collagen and fibronectin with fibrosis. A variety of organs, including kidney, liver, lung, heart and skin, can undergo fibrosis under the stimulation of exogenous or endogenous pathogenic factors. The orphan nuclear receptor 4 group A1 (NR4A1) and nuclear receptor 4 group A2(NR4A2)are belong to the nuclear receptor subfamily and inhibit the occurrence and development of fibrosis. NR4A1 is an inhibitory factor of TGF-ß signaling transduction. Overexpression of NR4A1 in fibroblasts can reduce TGF-ß induced collagen deposition and fibrosis related gene expression. Here, we summarize the current research progress on the NR4A1/2 and fibrosis, providing reference for the treatment of fibrosis.


Sujet(s)
Fibrose , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires , Humains , Animaux , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie
11.
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
12.
Arkh Patol ; 86(4): 58-63, 2024.
Article de Russe | MEDLINE | ID: mdl-39073544

RÉSUMÉ

A literature review reflects data on the mechanisms of pulmonary fibrosis after a novel coronavirus infection associated with the SARS-COV2 virus. Factors contributing to post-COVID lung remodeling are considered. According to the literature, in the mechanism of pulmonary fibrosis, during the course of the disease and during the recovery period, both direct viral damage and death of alveolocytes and endothelium, the development of a systemic inflammatory reaction due to inadequate secretion of cytokines, especially type 2, which are activators of the proliferation of fibroblasts and myofibroblasts, are important. The influence of angiogenesis disorders and vascular dysfunction on pneumofibrosis was noted. Attention is also paid to the relationship between the development of pulmonary fibrosis and abnormal activation of the renin-angiotensin-aldosterone system. In combination with the action of many factors, especially germinal ones, an imbalance between profibrogenic and antifibrogenic action develops and fibrosis occurs.


Sujet(s)
COVID-19 , Fibrose pulmonaire , SARS-CoV-2 , Humains , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/étiologie , Fibrose pulmonaire/métabolisme , COVID-19/complications , COVID-19/anatomopathologie , Système rénine-angiotensine , Cytokines/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Myofibroblastes/métabolisme
13.
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
14.
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
15.
Matrix Biol ; 132: 72-86, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39009171

RÉSUMÉ

Post-mitotic, non-proliferative dermal fibroblasts have crucial functions in maintenance and restoration of tissue homeostasis. They are involved in essential processes such as wound healing, pigmentation and hair growth, but also tumor development and aging-associated diseases. These processes are energetically highly demanding and error prone when mitochondrial damage occurs. However, mitochondrial function in fibroblasts and the influence of mitochondrial dysfunction on fibroblast-specific demands are still unclear. To address these questions, we created a mouse model in which accelerated cell-specific mitochondrial DNA (mtDNA) damage accumulates. We crossed mice carrying a dominant-negative mutant of the mitochondrial replicative helicase Twinkle (RosaSTOP system) with mice that express fibroblast-specific Cre Recombinase (Collagen1A2 CreERT) which can be activated by Tamoxifen (TwinkleFIBRO). Thus, we are able to induce mtDNA deletions and duplications in specific cells, a process which resembles the physiological aging process in humans, where this damage accumulates in all tissues. Upon proliferation in vitro, Tamoxifen induced Twinkle fibroblasts deplete most of their mitochondrial DNA which, although not disturbing the stoichiometry of the respiratory chain complexes, leads to reduced ROS production and mitochondrial membrane potential as well as an anti-inflammatory and anti-fibrotic profile of the cells. In Sodium Azide treated wildtype fibroblasts, without a functioning respiratory chain, we observe the opposite, a rather pro-inflammatory and pro-fibrotic signature. Upon accumulation of mitochondrial DNA mutations in vivo the TwinkleFIBRO mice are protected from fibrosis development induced by intradermal Bleomycin injections. This is due to dampened differentiation of the dermal fibroblasts into α-smooth-muscle-actin positive myofibroblasts in TwinkleFIBRO mice. We thus provide evidence for striking differences of the impact that mtDNA mutations have in contrast to blunted mitochondrial function in dermal fibroblasts and skin homeostasis. These data contribute to improved understanding of mitochondrial function and dysfunction in skin and provide mechanistic insight into potential targets to treat skin fibrosis in the future.


Sujet(s)
Bléomycine , Différenciation cellulaire , ADN mitochondrial , Fibrose , Mutation , Myofibroblastes , Animaux , Bléomycine/effets indésirables , Bléomycine/toxicité , ADN mitochondrial/génétique , ADN mitochondrial/métabolisme , Souris , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Myofibroblastes/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Helicase/génétique , Helicase/métabolisme , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Tamoxifène/pharmacologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/anatomopathologie , Modèles animaux de maladie humaine , Espèces réactives de l'oxygène/métabolisme , Humains , Peau/anatomopathologie , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Protéines mitochondriales/génétique , Protéines mitochondriales/métabolisme , Collagène de type I
16.
Matrix Biol ; 132: 87-97, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39019241

RÉSUMÉ

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) represents a severe and progressive manifestation of idiopathic interstitial pneumonia marked by an uncertain etiology along with an unfavorable prognosis. Osteoglycin (OGN), belonging to the small leucine-rich proteoglycans family, assumes pivotal functions in both tissue formation and damage response. However, the roles and potential mechanisms of OGN in the context of lung fibrosis remain unexplored. METHODS: The assessment of OGN expression levels in fibrotic lungs was conducted across various experimental lung fibrosis mouse models. To elucidate the effects of OGN on the differentiation of lung myofibroblasts, both OGN knockdown and OGN overexpression were employed in vitro. The expression of integrin αv, along with its colocalization with lysosomes and latency-associated peptide (LAP), was monitored in OGN-knockdown lung myofibroblasts. Furthermore, the role of OGN in lung fibrosis was investigated through OGN knockdown utilizing adeno-related virus serotype 6 (AAV6)-mediated delivery. RESULTS: OGN exhibited upregulation in both lungs and myofibroblasts across diverse lung fibrosis mouse models. And laboratory experiments in vitro demonstrated that OGN knockdown inhibited the TGF-ß/Smad signaling pathway in lung myofibroblasts. Conversely, OGN overexpression promoted TGF-ß/Smad pathway in these cells. Mechanistic insights revealed that OGN knockdown facilitated lysosome-mediated degradation of integrin αv while inhibiting its binding to latency-associated peptide (LAP). Remarkably, AAV6-targeted OGN knockdown ameliorated the extent of lung fibrosis in experimental mouse models. CONCLUSION: Our results indicate that inhibiting OGN signaling could serve as a promising therapeutic way for lung fibrosis.


Sujet(s)
Modèles animaux de maladie humaine , Fibrose pulmonaire idiopathique , Intégrine alphaV , Poumon , Myofibroblastes , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Souris , Intégrine alphaV/métabolisme , Intégrine alphaV/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance transformant bêta/génétique , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/génétique , Poumon/métabolisme , Poumon/anatomopathologie , Protéines Smad/métabolisme , Protéines Smad/génétique , Humains , Techniques de knock-down de gènes , Mâle , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/génétique , Fibrose pulmonaire/anatomopathologie
18.
Respir Res ; 25(1): 242, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38877465

RÉSUMÉ

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


Sujet(s)
Alcaloïdes , Différenciation cellulaire , Fibroblastes , Souris de lignée C57BL , Myofibroblastes , Fibrose pulmonaire , Silice , Silicose , Animaux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Alcaloïdes/pharmacologie , Silice/toxicité , Souris , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Silicose/anatomopathologie , Silicose/métabolisme , Silicose/traitement médicamenteux , Mâle
19.
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
20.
Life Sci ; 351: 122805, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38851422

RÉSUMÉ

AIMS: Heart failure (HF) is one of the most devastating consequences of cardiovascular diseases. Regardless of etiology, cardiac fibrosis is present and promotes the loss of heart function in HF patients. Cardiac resident fibroblasts, in response to a host of pro-fibrogenic stimuli, trans-differentiate into myofibroblasts to mediate cardiac fibrosis, the underlying mechanism of which remains incompletely understood. METHODS: Fibroblast-myofibroblast transition was induced in vitro by exposure to transforming growth factor (TGF-ß). Cardiac fibrosis was induced in mice by either transverse aortic constriction (TAC) or by chronic infusion with angiotensin II (Ang II). RESULTS: Through bioinformatic screening, we identified Kruppel-like factor 6 (KLF6) as a transcription factor preferentially up-regulated in cardiac fibroblasts from individuals with non-ischemic cardiomyopathy (NICM) compared to the healthy donors. Further analysis showed that nuclear factor kappa B (NF-κB) bound to the KLF6 promoter and mediated KLF6 trans-activation by pro-fibrogenic stimuli. KLF6 knockdown attenuated whereas KLF6 over-expression enhanced TGF-ß induced fibroblast-myofibroblast transition in vitro. More importantly, myofibroblast-specific KLF6 depletion ameliorated cardiac fibrosis and rescued heart function in mice subjected to the TAC procedure or chronic Ang II infusion. SIGNIFICANCE: In conclusion, our data support a role for KLF6 in cardiac fibrosis.


Sujet(s)
Fibroblastes , Fibrose , Facteur-6 de type krüppel , Souris de lignée C57BL , Myofibroblastes , Animaux , Facteur-6 de type krüppel/métabolisme , Facteur-6 de type krüppel/génétique , Fibrose/métabolisme , Souris , Humains , Mâle , Fibroblastes/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Angiotensine-II/pharmacologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Facteur de croissance transformant bêta/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Cellules cultivées , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Défaillance cardiaque/métabolisme , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE