Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.544
Filtrer
1.
Hum Genomics ; 18(1): 80, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39014455

RÉSUMÉ

BACKGROUND: Keloid is a disease characterized by proliferation of fibrous tissue after the healing of skin tissue, which seriously affects the daily life of patients. However, the clinical treatment of keloids still has limitations, that is, it is not effective in controlling keloids, resulting in a high recurrence rate. Thus, it is urgent to identify new signatures to improve the diagnosis and treatment of keloids. METHOD: Bulk RNA seq and scRNA seq data were downloaded from the GEO database. First, we used WGCNA and MEGENA to co-identify keloid/immune-related DEGs. Subsequently, we used three machine learning algorithms (Randomforest, SVM-RFE, and LASSO) to identify hub immune-related genes of keloid (KHIGs) and investigated the heterogeneous expression of KHIGs during fibroblast subpopulation differentiation using scRNA-seq. Finally, we used HE and Masson staining, quantitative reverse transcription-PCR, western blotting, immunohistochemical, and Immunofluorescent assay to investigate the dysregulated expression and the mechanism of retinoic acid in keloids. RESULTS: In the present study, we identified PTGFR, RBP5, and LIF as KHIGs and validated their diagnostic performance. Subsequently, we constructed a novel artificial neural network molecular diagnostic model based on the transcriptome pattern of KHIGs, which is expected to break through the current dilemma faced by molecular diagnosis of keloids in the clinic. Meanwhile, the constructed IG score can also effectively predict keloid risk, which provides a new strategy for keloid prevention. Additionally, we observed that KHIGs were also heterogeneously expressed in the constructed differentiation trajectories of fibroblast subtypes, which may affect the differentiation of fibroblast subtypes and thus lead to dysregulation of the immune microenvironment in keloids. Finally, we found that retinoic acid may treat or alleviate keloids by inhibiting RBP5 to differentiate pro-inflammatory fibroblasts (PIF) to mesenchymal fibroblasts (MF), which further reduces collagen secretion. CONCLUSION: In summary, the present study provides novel immune signatures (PTGFR, RBP5, and LIF) for keloid diagnosis and treatment, and identifies retinoic acid as potential anti-keloid drugs. More importantly, we provide a new perspective for understanding the interactions between different fibroblast subtypes in keloids and the remodeling of their immune microenvironment.


Sujet(s)
Chéloïde , RNA-Seq , Chéloïde/génétique , Chéloïde/diagnostic , Chéloïde/anatomopathologie , Chéloïde/immunologie , Chéloïde/traitement médicamenteux , Humains , Transcriptome/génétique , Analyse de profil d'expression de gènes , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Réseaux de régulation génique , Trétinoïne/pharmacologie , Trétinoïne/usage thérapeutique , Analyse sur cellule unique/méthodes , Différenciation cellulaire/génétique , Analyse de séquence d'ARN/méthodes , Apprentissage machine , Analyse de l'expression du gène de la cellule unique
2.
PLoS One ; 19(7): e0305927, 2024.
Article de Anglais | MEDLINE | ID: mdl-39024326

RÉSUMÉ

Fibrotic skin diseases, such as keloids, are pathological results of aberrant tissue healing and are characterized by overgrowth of dermal fibroblasts. Remdesivir (RD), an antiviral drug, has been reported to have pharmacological activities in a wide range of fibrotic diseases. However, whether RD function on skin fibrosis remains unclear. Therefore, in our study, we explored the potential effect and mechanisms of RD on skin fibrosis both in vivo and in vitro. As expected, the results demonstrated that RD alleviated BLM-induced skin fibrosis and attenuates the gross weight of keloid tissues in vivo. Further studies suggested that RD suppressed fibroblast activation and autophagy both in vivo and in vitro. In addition, mechanistic research showed that RD attenuated fibroblasts activation by the TGF-ß1/Smad signaling pathway and inhibited fibroblasts autophagy by the PI3K/Akt/mTOR signaling pathway. In summary, our results demonstrate therapeutic potential of RD for skin fibrosis in the future.


Sujet(s)
AMP , Alanine , Fibroblastes , Fibrose , Transduction du signal , Peau , Facteur de croissance transformant bêta-1 , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-1/métabolisme , Fibrose/traitement médicamenteux , Alanine/analogues et dérivés , Alanine/pharmacologie , Alanine/usage thérapeutique , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , AMP/analogues et dérivés , AMP/pharmacologie , AMP/métabolisme , Souris , Peau/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/métabolisme , Humains , Autophagie/effets des médicaments et des substances chimiques , Chéloïde/traitement médicamenteux , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Antiviraux/pharmacologie , Sérine-thréonine kinases TOR/métabolisme , Bléomycine , Phosphatidylinositol 3-kinases/métabolisme , Mâle , Protéines proto-oncogènes c-akt/métabolisme , Protéines Smad/métabolisme
3.
Pathol Oncol Res ; 30: 1611789, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903488

RÉSUMÉ

Background: The desmoplastic reaction is considered a promising prognostic parameter for colorectal cancer. However, intermediate desmoplastic reaction is characterized by sizeable stromal heterogeneity, including both small amounts of keloid-like collagen (KC) in the fibrotic stroma and thick tufts of KC circumferentially surrounding cancer nests and occupying most of the fields of view. The present study aimed to evaluate the diagnostic and prognostic significance of KC histophenotyping with a quantitative visual assessment of its presence in the stroma of the invasive margin of TNM (The "tumor-node-metastasis" classification) stage II/III colorectal cancer (CRC). Methods and results: 175 resected tumors from patients with TNM stage II/III CRC were examined. Keloid-like collagen was assessed according to Ueno H. criteria. KC was assessed at the primary tumor invasive margin using Hematoxylin & Eosin and Masson's trichrome staining. The cut-off point for KC was examined using "the best cutoff approach by log-rank test." Using a cutoff point of 30%, we histologically divided fibrous stroma in the invasive area into two groups: "type A"-KC ≤ 0.3 and "type B"-KC>0.3. Type A stroma was observed in 48% of patients, type B-in 52%. The association between collagen amount and 5-year recurrence-free survival (5-RFS) was assessed using Cox regression analysis. Kaplan-Meier analysis and log-rank tests were used to assess the significance of survival analysis. Analysis of categorical variables showed that increased KC in CRC stroma predicted adverse outcomes for 5-RFS (hazard ratio [HR] = 3.143, 95%, confidence interval [CI] = 1.643-6.012, p = 0.001). Moreover, in Kaplan-Meier analysis, the log-rank test showed that type B exhibited worse 5-RFS than type A (p = 0.000). Conclusion: KC is an independent predictor of 5-year overall and RFS in patients with TNM stage II/III CRC treated with surgery, with worse survival rates when the amount of KC increases by >30%.


Sujet(s)
Collagène , Tumeurs colorectales , Matrice extracellulaire , Chéloïde , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Mâle , Femelle , Pronostic , Adulte d'âge moyen , Collagène/métabolisme , Sujet âgé , Matrice extracellulaire/anatomopathologie , Matrice extracellulaire/métabolisme , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Adulte , Sujet âgé de 80 ans ou plus , Taux de survie , Études de suivi
4.
Exp Dermatol ; 33(6): e15111, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38840411

RÉSUMÉ

Keloids are pathological scar tissue resulting from skin trauma or spontaneous formation, often accompanied by itching and pain. Although GNAS antisense RNA 1 (GNAS-AS1) shows abnormal upregulation in keloids, the underlying molecular mechanism is unclear. The levels of genes and proteins in clinical tissues from patients with keloids and human keloid fibroblasts (HKFs) were measured using quantitative reverse transcription PCR, western blot and enzyme-linked immunosorbent assay. The features of HKFs, including proliferation and migration, were evaluated using cell counting kit 8 and a wound healing assay. The colocalization of GNAS-AS1 and miR-196a-5p in HKFs was measured using fluorescence in situ hybridization. The relationships among GNAS-AS1, miR-196a-5p and C-X-C motif chemokine ligand 12 (CXCL12) in samples from patients with keloids were analysed by Pearson correlation analysis. Gene interactions were validated by chromatin immunoprecipitation and luciferase reporter assays. GNAS-AS1 and CXCL12 expression were upregulated and miR-196a-5p expression was downregulated in clinical tissues from patients with keloids. GNAS-AS1 knockdown inhibited proliferation, migration, and extracellular matrix (ECM) accumulation of HKFs, all of which were reversed by miR-196a-5p downregulation. Signal transducer and activator of transcription 3 (STAT3) induced GNAS-AS1 transcription through GNAS-AS1 promoter interaction, and niclosamide, a STAT3 inhibitor, decreased GNAS-AS1 expression. GNAS-AS1 positively regulated CXCL12 by sponging miR-196-5p. Furthermore, CXCL12 knockdown restrained STAT3 phosphorylation in HKFs. Our findings revealed a feedback loop of STAT3/GNAS-AS1/miR-196a-5p/CXCL12/STAT3 that promoted HKF proliferation, migration and ECM accumulation and affected keloid progression.


Sujet(s)
Prolifération cellulaire , Chimiokine CXCL12 , Fibroblastes , Chéloïde , microARN , ARN long non codant , Facteur de transcription STAT-3 , Chéloïde/métabolisme , Chéloïde/génétique , Chéloïde/anatomopathologie , Humains , microARN/métabolisme , microARN/génétique , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Chimiokine CXCL12/métabolisme , Chimiokine CXCL12/génétique , Fibroblastes/métabolisme , Mouvement cellulaire , Rétrocontrôle physiologique , Chromogranine/génétique , Chromogranine/métabolisme , Mâle , Femelle , Sous-unités alpha Gs des protéines G/génétique , Sous-unités alpha Gs des protéines G/métabolisme , Transduction du signal , Adulte , Cellules cultivées , Régulation positive
5.
Arch Dermatol Res ; 316(7): 412, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38878082

RÉSUMÉ

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


Sujet(s)
Prolifération cellulaire , Fibroblastes , Folliculite , Glycolyse , Chéloïde , Humains , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Folliculite/métabolisme , Folliculite/anatomopathologie , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Cellules cultivées , Phosphorylation oxydative , Mouvement cellulaire , Adulte , Peau/anatomopathologie , Peau/métabolisme , Métabolisme énergétique , Femelle , Mâle
6.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892032

RÉSUMÉ

Keloids, marked by abnormal cellular proliferation and excessive extracellular matrix (ECM) accumulation, pose significant therapeutic challenges. Ethyl pyruvate (EP), an inhibitor of the high-mobility group box 1 (HMGB1) and TGF-ß1 pathways, has emerged as a potential anti-fibrotic agent. Our research evaluated EP's effects on keloid fibroblast (KF) proliferation and ECM production, employing both in vitro cell cultures and ex vivo patient-derived keloid spheroids. We also analyzed the expression levels of ECM components in keloid tissue spheroids treated with EP through immunohistochemistry. Findings revealed that EP treatment impedes the nuclear translocation of HMGB1 and diminishes KF proliferation. Additionally, EP significantly lowered mRNA and protein levels of collagen I and III by attenuating TGF-ß1 and pSmad2/3 complex expression in both human dermal fibroblasts and KFs. Moreover, metalloproteinase I (MMP-1) and MMP-3 mRNA levels saw a notable increase following EP administration. In keloid spheroids, EP induced a dose-dependent reduction in ECM component expression. Immunohistochemical and western blot analyses confirmed significant declines in collagen I, collagen III, fibronectin, elastin, TGF-ß, AKT, and ERK 1/2 expression levels. These outcomes underscore EP's antifibrotic potential, suggesting its viability as a therapeutic approach for keloids.


Sujet(s)
Fibroblastes , Chéloïde , Pyruvates , Sphéroïdes de cellules , Humains , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Pyruvates/pharmacologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Facteur de croissance transformant bêta-1/métabolisme , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Collagène/métabolisme , Collagène/biosynthèse , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Matrix metalloproteinase 3/métabolisme , Matrix metalloproteinase 3/génétique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Collagène de type I/métabolisme , Collagène de type I/génétique , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Protéine Smad-3/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Mâle
7.
Front Immunol ; 15: 1326728, 2024.
Article de Anglais | MEDLINE | ID: mdl-38915394

RÉSUMÉ

Keloids are a common connective tissue disorder with an ill-understood etiopathogenesis and no effective treatment. This is exacerbated because of the absence of an animal model. Patient-derived primary keloid cells are insufficient as they age through passaging and have a limited supply. Therefore, there is an unmet need for development of a cellular model that can consistently and faithfully represent keloid's pathognomic features. In view of this, we developed keloid-derived immortalized fibroblast (KDIF) cell lines from primary keloid fibroblasts (PKF) by transfecting the human telomerase reverse transcriptase (hTERT) gene. The TERT gene encodes the catalytic subunit of the telomerase enzyme, which is responsible for maintaining the cellular replicative potential (cellular immortalization). Primary fibroblasts from keloid-specific lesional (peripheral, middle, and top) as well as extralesional sites were isolated and evaluated for cell line development and comparative cellular characteristics by employing qRT-PCR and immunofluorescence staining. Moreover, the immortalized behavior of KDIF cell lines was evaluated by comparing with cutaneous fibrosarcoma and dermatofibrosarcoma protuberans cell lines. Stable KDIF cell lines with elevated expression of hTERT exhibited the cellular characteristics of site-specific keloid fibroblasts. Histochemical staining for ß-galactosidase revealed a significantly lower number of ß-gal-positive cells in all three KDIF cell lines compared with that in PKFs. The cell growth curve pattern was studied over 10 passages for all three KDIF cell lines and was compared with the control groups. The results showed that all three KDIF cell lines grew significantly faster and obtained a fast growing characteristic as compared to primary keloid and normal fibroblasts. Phenotypic behavior in growth potential is an indication of hTERT-mediated immortalized transformation. Cell migration analysis revealed that the top and middle KDIF cell lines exhibited similar migration trend as site-specific PKFs. Notably, peripheral KDIF cell line showed significantly enhanced cell migration in comparison to the primary peripheral fibroblasts. All KDIF cell lines expressed Collagen I protein as a keloid-associated fibrotic marker. Functional testing with triamcinolone inhibited cell migration in KDIF. ATCC short tandem repeat profiling validated the KDIF as keloid representative cell line. In summary, we provide the first novel KDIF cell lines. These cell lines overcome the limitations related to primary cell passaging and tissue supply due to immortalized features and present an accessible and consistent experimental model for keloid research.


Sujet(s)
Fibroblastes , Chéloïde , Telomerase , Humains , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Fibroblastes/métabolisme , Telomerase/génétique , Telomerase/métabolisme , Lignée cellulaire , Lignée de cellules transformées , Mâle , Femelle , Adulte , Adulte d'âge moyen
8.
Toxicol Appl Pharmacol ; 489: 117012, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38906511

RÉSUMÉ

Keloid formation has been linked to abnormal fibroblast function, such as excessive proliferation and extracellular matrix (ECM) production. Serum deprivation protein response (SDPR) is a crucial regulator of cellular function under diverse pathological conditions, yet its role in keloid formation remains unknown. The current work investigated the function of SDPR in regulating the proliferation, motility, and ECM production of keloid fibroblasts (KFs), as well as to decipher the mechanisms involved. Analysis of RNA sequencing data from the GEO database demonstrated significant down-regulation of SDPR in KF compared to normal fibroblasts (NFs). This down-regulation was also observed in clinical keloid specimens and isolated KFs. Overexpression of SDPR suppressed the proliferation, motility, and ECM production of KFs, while depletion of SDPR exacerbated the enhancing impact of TGF-ß1 on the proliferation, motility, and ECM production of NFs. Mechanistic studies revealed that SDPR overexpression repressed TGF-ß/Smad signal cascade activation in KFs along with decreased levels of phosphorylated Samd2/3, while SDPR depletion exacerbated TGF-ß/Smad activation in TGF-ß1-stimulated NFs. SDPR overexpression also repressed ERK1/2 activation in KFs, while SDPR depletion exacerbated ERK1/2 activation in TGF-ß1-stimulated NFs. Inhibition of ERK1/2 abolished SDPR-depletion-induced TGF-ß1/Smad activation, cell proliferation, motility, and ECM production in NFs. In conclusion, SDPR represses the proliferation, motility, and ECM production in KFs by blocking the TGF-ß1/Smad pathway in an ERK1/2-dependent manner. The findings highlight the role of SDPR in regulating abnormal behaviors of fibroblasts associated with keloid formation and suggest it as a potential target for anti-keloid therapy development.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Matrice extracellulaire , Fibroblastes , Chéloïde , Système de signalisation des MAP kinases , Protéines Smad , Facteur de croissance transformant bêta-1 , Humains , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Chéloïde/génétique , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-1/métabolisme , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Protéines Smad/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/physiologie , Transduction du signal , Cellules cultivées , Mitogen-Activated Protein Kinase 1/métabolisme , Mâle , Femelle , Mitogen-Activated Protein Kinase 3/métabolisme , Adulte
9.
Int Immunopharmacol ; 137: 112423, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38861914

RÉSUMÉ

Fibrosis is the excessive deposition of extracellular matrix in an organ or tissue that results from an impaired tissue repair in response to tissue injury or chronic inflammation. The progressive nature of fibrotic diseases and limited treatment options represent significant healthcare challenges. Despite the substantial progress in understanding the mechanisms of fibrosis, a gap persists translating this knowledge into effective therapeutics. Here, we discuss the critical mediators involved in fibrosis and the role of tranilast as a potential antifibrotic drug to treat fibrotic conditions. Tranilast, an antiallergy drug, is a derivative of tryptophan and has been studied for its role in various fibrotic diseases. These include scleroderma, keloid and hypertrophic scars, liver fibrosis, renal fibrosis, cardiac fibrosis, pulmonary fibrosis, and uterine fibroids. Tranilast exerts antifibrotic effects by suppressing fibrotic pathways, including TGF-ß, and MPAK. Because it disrupts fibrotic pathways and has demonstrated beneficial effects against keloid and hypertrophic scars, tranilast could be used to treat other conditions characterized by fibrosis.


Sujet(s)
Fibrose , Transduction du signal , ortho-Aminobenzoates , Humains , ortho-Aminobenzoates/usage thérapeutique , ortho-Aminobenzoates/pharmacologie , Fibrose/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Antifibrotiques/usage thérapeutique , Antifibrotiques/pharmacologie , Chéloïde/traitement médicamenteux , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Facteur de croissance transformant bêta/métabolisme
10.
J Cell Biochem ; 125(7): e30609, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38860429

RÉSUMÉ

Keloid is a typical fibrotic and inflammatory skin disease with unclear mechanisms and few therapeutic targets. In this study, we found that BMP1 was significantly increased in a collagen high-expressing subtype of fibroblast by reanalyzing a public single-cell RNA-sequence data set of keloid. The number of BMP1-positive fibroblast cells was increased in keloid fibrotic loci. Increased levels of BMP1 were further validated in the skin tissues and fibroblasts from keloid patients. Additionally, a positive correlation between BMP1 and the Keloid Area and Severity Index was found in keloid patients. In vitro analysis revealed collagen production, the phosphorylation levels of p65, and the IL-1ß secretion decreased in BMP1 interfered keloid fibroblasts. Besides, the knockdown of BMP1 inhibited the growth and migration of keloid fibroblast cells. Mechanistically, BMP1 inhibition downregulated the noncanonical TGF-ß pathways, including p-p38 and p-ERK1/2 signaling. Furthermore, we found the delivery of BMP1 siRNAs could significantly alleviate keloid in human keloid-bearing nude mice. Collectively, our results indicated that BMP1 exhibited various pathogenic effects on keloids as promoting cell proliferation, migration, inflammation, and ECM deposition of fibroblast cells by regulating the noncanonical TGF-ß/p38 MAPK, and TGF-ß/ERK pathways. BMP1-lowing strategies may appear as a potential new therapeutic target for keloid.


Sujet(s)
Protéine morphogénétique osseuse de type 1 , Fibroblastes , Inflammation , Chéloïde , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Chéloïde/génétique , Humains , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Protéine morphogénétique osseuse de type 1/métabolisme , Protéine morphogénétique osseuse de type 1/génétique , Animaux , Souris , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Mâle , Souris nude , Prolifération cellulaire , Femelle , Mouvement cellulaire , Fibrose , Adulte , Facteur de croissance transformant bêta/métabolisme , Système de signalisation des MAP kinases
11.
PeerJ ; 12: e17551, 2024.
Article de Anglais | MEDLINE | ID: mdl-38887622

RÉSUMÉ

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


Sujet(s)
Cyclohexylamines , Ferroptose , Fibroblastes , Fibrose , Chéloïde , Facteur-2 apparenté à NF-E2 , Phénylènediamines , Phospholipid hydroperoxide glutathione peroxidase , Humains , Ferroptose/effets des médicaments et des substances chimiques , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Chéloïde/traitement médicamenteux , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Cyclohexylamines/pharmacologie , Fibrose/métabolisme , Fibrose/anatomopathologie , Phénylènediamines/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Mâle , Peroxydation lipidique/effets des médicaments et des substances chimiques , Femelle , Adulte , Fer/métabolisme , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Pipérazines/pharmacologie , Actines/métabolisme , Actines/génétique , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques
12.
Arch Dermatol Res ; 316(7): 368, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38850361

RÉSUMÉ

Intralesional corticosteroid injections are a first-line treatment for keloids; yet clinical treatment results are highly variable and often suboptimal. Variation in triamcinolone acetonide (TAC) biodistribution may be an important reason for the variable effects of TAC treatment in keloids. In this exploratory study we investigated the biodistribution of TAC in keloids and normal skin using different drug delivery techniques. Fluorescent-labeled TAC suspension was administered into keloids and normal skin with a hypodermic needle and an electronic pneumatic jet injector. TAC biodistribution was represented by the fluorescent TAC volume and 3D biodistribution shape of TAC, using a 3D-Fluorescence-Imaging Cryomicrotome System. Twenty-one keloid and nine normal skin samples were analyzed. With needle injections, the mean fluorescent TAC volumes were 990 µl ± 479 in keloids and 872 µl ± 227 in normal skin. With the jet injector, the mean fluorescent TAC volumes were 401 µl ± 252 in keloids and 249 µl ± 67 in normal skin. 3D biodistribution shapes of TAC were highly variable in keloids and normal skin. In conclusion, TAC biodistribution in keloids is highly variable for both needle and jet injection. This may partly explain the variable treatment effects of intralesional TAC in keloids. Future research is needed to confirm this preliminary finding and to optimize drug delivery in keloids.


Sujet(s)
Chéloïde , Triamcinolone acétonide , Chéloïde/traitement médicamenteux , Chéloïde/anatomopathologie , Humains , Triamcinolone acétonide/pharmacocinétique , Triamcinolone acétonide/administration et posologie , Adulte , Femelle , Distribution tissulaire , Mâle , Adulte d'âge moyen , Injections intralésionnelles , Peau/métabolisme , Peau/anatomopathologie , Peau/imagerie diagnostique , Cryo-ultramicrotomie/méthodes , Jeune adulte , Imagerie tridimensionnelle , Systèmes de délivrance de médicaments/méthodes
13.
Int J Mol Sci ; 25(10)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38791322

RÉSUMÉ

A keloid is a benign fibroproliferative hypertrophy of scar tissue that extends outside the original wound and invades adjacent healthy skin. Keloid formation is thought to be a complex process including overactivity of the interleukin-6 signaling pathway and genetic susceptibility. The aim of the study was to investigate possible associations between rs1800797, rs1800796, and rs1800795 polymorphisms in the promoter of the IL6 gene encoding interleukin-6 and the rs2228145 polymorphism in the IL6R gene encoding the interleukin-6 receptor subunit alpha with the predisposition to keloids in Polish patients. The genetic polymorphisms were identified either using Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP) or sequencing of samples of genomic DNA extracted from blood leukocytes of 86 adult patients with keloids and 100 newborns comprising a control group. No significant differences in the distributions of IL6 or IL6R alleles or genotypes were found between keloid patients and newborn controls. There were also no significant differences between both groups in the distribution of IL6 haplotypes. The IL6 rs1800797, rs1800796 and rs1800795 and IL6R rs2228145 polymorphisms were not found to predispose individuals in the study group to keloids. IL6 promoter haplotypes were not found to be associated with a higher risk of keloids in the studied group.


Sujet(s)
Prédisposition génétique à une maladie , Interleukine-6 , Chéloïde , Polymorphisme de nucléotide simple , Récepteurs à l'interleukine-6 , Humains , Chéloïde/génétique , Chéloïde/anatomopathologie , Interleukine-6/génétique , Récepteurs à l'interleukine-6/génétique , Mâle , Femelle , Adulte , Pologne , Adulte d'âge moyen , Régions promotrices (génétique) , Études cas-témoins , Haplotypes , Allèles , Adolescent , Jeune adulte , Fréquence d'allèle , Génotype , Nouveau-né , Études d'associations génétiques
14.
Article de Chinois | MEDLINE | ID: mdl-38664034

RÉSUMÉ

Skin fibrosis diseases mainly include hypertrophic scar, keloid, and systemic sclerosis, etc. The main pathological features are excessive activation of fibroblasts and abnormal deposition of extracellular matrix. In recent years, studies have shown that aerobic glycolysis is closely related to the occurrence and development of skin fibrosis diseases. Drugs targeting aerobic glycolysis has provided new ideas for skin anti-fibrosis treatment. This article reviews the role of enzymes and products related to aerobic glycolysis in the occurrence and development of skin fibrosis diseases and the drugs targeting aerobic glycolysis for the treatment of skin fibrosis diseases.


Sujet(s)
Fibrose , Glycolyse , Humains , Fibrose/métabolisme , Fibrose/anatomopathologie , Maladies de la peau/métabolisme , Maladies de la peau/anatomopathologie , Maladies de la peau/traitement médicamenteux , Peau/anatomopathologie , Peau/métabolisme , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Chéloïde/traitement médicamenteux , Sclérodermie systémique/métabolisme , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/traitement médicamenteux
15.
Biochem Biophys Res Commun ; 715: 149963, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38676999

RÉSUMÉ

Keloids represent a prevalent dermal fibroproliferative disorder. They only affect humans and exhibit several tumor characteristics, such as excessive extracellular matrix (ECM) deposition, which usually occurs after skin injury. Kreotoxin type A (KTA) can inhibit the release of acetylcholine, and thereby inhibit the proliferation of keloid fibroblasts and reducing the formation of scars. Thus, KTA could be used as a therapeutic agent for keloids. However, the mechanisms of action of KTA in keloid treatment remain unclear. In this study, we aimed to explore the underlying mechanisms of action of KTA in human keloid treatment using human tissue and a cell-based model. Integrative microarray analysis revealed that hypoxia-inducible factor 1-alpha (HIF-1α) expression was frequently upregulated in hypertrophic scar and keloid tissues, whereas it was downregulated in the KTA-treated samples. Furthermore, KTA addition to keloid-derived fibroblasts (KDFs) reduced the growth rate and viability, induced apoptosis, and decreased inflammation and oxidative stress in KDFs. However, overexpression of HIF-1α restored cell number and survival, decreased apoptosis, and promoted inflammation and oxidative stress in KTA-treated KDFs. Furthermore, KTA treatment reduced the expression of ECM proteins, including vascular endothelial growth factor (VEGF), collagen I and III, whereas HIF-1α overexpression abolished the effects of KTA on KDFs. In conclusion, our findings provide novel insights into the mechanisms of action of KTA as a potential therapeutic agent for keloids via modulating HIF-1α expression.


Sujet(s)
Prolifération cellulaire , Régulation négative , Fibroblastes , Sous-unité alpha du facteur-1 induit par l'hypoxie , Inflammation , Chéloïde , Humains , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Régulation négative/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/effets des médicaments et des substances chimiques , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Toxines bactériennes/pharmacologie
16.
Wound Repair Regen ; 32(4): 419-428, 2024.
Article de Anglais | MEDLINE | ID: mdl-38602106

RÉSUMÉ

Keloid scars tend to occur in high-tension sites due to mechanical stimuli that are involved in their development. To date, a detailed analysis of keloid distribution focused specifically on facial and neck areas has not been reported, and limited literature exists as to the related mechanical factors. To rectify this deficiency of knowledge, we first quantified the facial and neck keloid distribution observed clinically in 113 patients. Subsequently, we performed a rigorous investigation into the mechanical factors and their associated changes at these anatomic sites in healthy volunteers without a history of pathologic scarring. The association between keloid-predilection sites and sebaceous gland-dense and acne-prone sites was also examined. To assess skin stretch, thickness and stiffness, VECTRA, ultrasound and indentometer were utilised. Baseline skin stiffness and thickness were measured, as well as the magnitude of change in these values associated with facial expression and postural changes. Within the face and neck, keloids were most common near the mandibular angle (41.3%) and lateral submental (20.0%) regions. These areas of increased keloid incidence were not associated with areas more dense in sebaceous glands, nor linked consistently with acne-susceptible regions. Binomial logistic regression revealed that changes in skin stiffness and thickness related to postural changes significantly predicted keloid distribution. Skin stiffness and thickness changes related to prolonged mechanical forces (postural changes) are most pronounced at sites of high keloid predilection. This finding further elucidates the means by which skin stretch and tension are related to keloid development. As a more detailed analysis of mechanical forces on facial and neck skin, this study evaluates the nuances of multiple skin-mechanical properties, and their changes in a three-dimensional framework. Such factors may be critical to better understanding keloid progression and development in the face and neck.


Sujet(s)
Face , Chéloïde , Cou , Peau , Humains , Chéloïde/anatomopathologie , Chéloïde/physiopathologie , Mâle , Femelle , Cou/anatomopathologie , Face/anatomopathologie , Adulte , Peau/anatomopathologie , Adulte d'âge moyen , Mouvement/physiologie , Jeune adulte , Adolescent
18.
J Gene Med ; 26(5): e3688, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38686583

RÉSUMÉ

BACKGROUND: Despite the interest in mesenchymal stem cells (MSC), their potential to treat abnormal scarring, especially keloids, is yet to be described. The present study aimed to investigate the therapeutic potential of exosomes derived from human bone marrow MSCs (hBMSC-Exos) in alleviating keloid formation. METHODS: Exosomes were isolated from hBMSC, and keloid fibroblasts (KFs) were treated with hBMSC-Exos. Cell counting kit-8, wound healing, transwell invasion, immunofluorescence, and western blot assays were conducted to study the malignant phenotype of KFs. Mice were induced with keloids and treated with hBMSC-Exos. The effect of hBMSC-Exos on keloid formation in vivo was evaluated by hematoxylin and eosin staining, Masson staining, immunohistochemistry, and western blotting. The GSE182192 dataset was screened for differentially expressed long non-coding RNA during keloid formation. Next, maternally expressed gene 3 (MEG3) was knocked down in hBMSC to obtain hBMSC-Exossh-MEG3. The molecular mechanism of MEG3 was investigated by bioinformatic screening, and the relationship between MEG3 and TP53 or MCM5 was verified. RESULTS: hBMSC-Exos inhibited the malignant proliferation, migration, and invasion of KFs at same time as promoting their apoptosis, Moreover, hBMSC-Exos reduced the expression of fibrosis- and collagen-related proteins in the cells and the formation of keloids caused by KFs. The reduction in MEG3 enrichment in hBMSC-Exos weakened the inhibitory effect of hBMSC-Exos on KF activity. hBMSC-Exos delivered MEG3 to promote MCM5 transcription by TP53 in KFs. Overexpression of MCM5 in KFs reversed the effects of hBMSC-Exossh-MEG3, leading to reduced KF activity. CONCLUSIONS: hBMSC-Exos delivered MEG3 to promote the protein stability of TP53, thereby activating MCM5 and promoting KF activity.


Sujet(s)
Exosomes , Fibroblastes , Chéloïde , Cellules souches mésenchymateuses , ARN long non codant , Protéine p53 suppresseur de tumeur , Animaux , Femelle , Humains , Mâle , Souris , Prolifération cellulaire , Modèles animaux de maladie humaine , Exosomes/métabolisme , Exosomes/génétique , Fibroblastes/métabolisme , Régulation de l'expression des gènes , Chéloïde/métabolisme , Chéloïde/génétique , Chéloïde/anatomopathologie , Chéloïde/thérapie , Cellules souches mésenchymateuses/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique
19.
Exp Dermatol ; 33(5): e15088, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38685820

RÉSUMÉ

Recently, the pathomechanisms of keloids have been extensively researched using transcriptomic analysis, but most studies did not consider the activity of keloids. We aimed to profile the transcriptomics of keloids according to their clinical activity and location within the keloid lesion, compared with normal and mature scars. Tissue samples were collected (keloid based on its activity (active and inactive), mature scar from keloid patients and normal scar (NS) from non-keloid patients). To reduce possible bias, all keloids assessed in this study had no treatment history and their location was limited to the upper chest or back. Multiomics assessment was performed by using single-cell RNA sequencing and multiplex immunofluorescence. Increased mesenchymal fibroblasts (FBs) was the main feature in keloid patients. Noticeably, the proportion of pro-inflammatory FBs was significantly increased in active keloids compared to inactive ones. To explore the nature of proinflammatory FBs, trajectory analysis was conducted and CCN family associated with mechanical stretch exhibited higher expression in active keloids. For vascular endothelial cells (VECs), the proportion of tip and immature cells increased in keloids compared to NS, especially at the periphery of active keloids. Also, keloid VECs highly expressed genes with characteristics of mesenchymal activation compared to NS, especially those from the active keloid center. Multiomics analysis demonstrated the distinct expression profile of active keloids. Clinically, these findings may provide the future appropriate directions for development of treatment modalities of keloids. Prevention of keloids could be possible by the suppression of mesenchymal activation between FBs and VECs and modulation of proinflammatory FBs may be the key to the control of active keloids.


Sujet(s)
Fibroblastes , Chéloïde , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Humains , Fibroblastes/métabolisme , Transcriptome , Cellules endothéliales/métabolisme , Femelle , Adulte , Mâle , Analyse de profil d'expression de gènes , Analyse sur cellule unique
20.
Matrix Biol ; 128: 79-92, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38485100

RÉSUMÉ

Keloid refers to a fibroproliferative disorder characterized by an accumulation of extracellular matrix (ECM) components at the dermis level, overgrowth beyond initial wound, and formation of tumor-like nodule areas. Treating keloid is still an unmet clinical need and the lack of an efficient therapy is clearly related to limited knowledge about keloid etiology, despite the growing interest of the scientific community in this pathology. In past decades, keloids were often studied in vitro through the sole prism of fibroblasts considered as the major effector of ECM deposition. Nevertheless, development of keloids results from cross-interactions of keloid fibroblasts (KFs) and their surrounding microenvironment, including immune cells such as macrophages. Our study aimed to evaluate the effect of M1 and M2 monocyte-derived macrophages on KFs in vitro. We focused on the effects of the macrophage secretome on fibrosis-related criteria in KFs, including proliferation, migration, differentiation, and ECM synthesis. First, we demonstrated that M2-like macrophages enhanced the fibrogenic profile of KFs in culture. Then, we surprisingly founded that M1-like macrophages can have an anti-fibrogenic effect on KFs, even in a pro-fibrotic environment. These results demonstrate, for the first time, that M1 and M2 macrophage subsets differentially impact the fibrotic fate of KFs in vitro, and suggest that restoring the M1/M2 balance to favor M1 in keloids could be an efficient therapeutic lever to prevent or treat keloid fibrosis.


Sujet(s)
Chéloïde , Humains , Chéloïde/génétique , Chéloïde/anatomopathologie , Fibroblastes/anatomopathologie , Prolifération cellulaire , Cellules cultivées
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE