Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 56.521
Filtrer
1.
Mol Cancer ; 23(1): 157, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095854

RÉSUMÉ

BACKGROUND: Tumor heterogeneity presents a formidable challenge in understanding the mechanisms driving tumor progression and metastasis. The heterogeneity of hepatocellular carcinoma (HCC) in cellular level is not clear. METHODS: Integration analysis of single-cell RNA sequencing data and spatial transcriptomics data was performed. Multiple methods were applied to investigate the subtype of HCC tumor cells. The functional characteristics, translation factors, clinical implications and microenvironment associations of different subtypes of tumor cells were analyzed. The interaction of subtype and fibroblasts were analyzed. RESULTS: We established a heterogeneity landscape of HCC malignant cells by integrated 52 single-cell RNA sequencing data and 5 spatial transcriptomics data. We identified three subtypes in tumor cells, including ARG1+ metabolism subtype (Metab-subtype), TOP2A+ proliferation phenotype (Prol-phenotype), and S100A6+ pro-metastatic subtype (EMT-subtype). Enrichment analysis found that the three subtypes harbored different features, that is metabolism, proliferating, and epithelial-mesenchymal transition. Trajectory analysis revealed that both Metab-subtype and EMT-subtype originated from the Prol-phenotype. Translation factor analysis found that EMT-subtype showed exclusive activation of SMAD3 and TGF-ß signaling pathway. HCC dominated by EMT-subtype cells harbored an unfavorable prognosis and a deserted microenvironment. We uncovered a positive loop between tumor cells and fibroblasts mediated by SPP1-CD44 and CCN2/TGF-ß-TGFBR1 interaction pairs. Inhibiting CCN2 disrupted the loop, mitigated the transformation to EMT-subtype, and suppressed metastasis. CONCLUSION: By establishing a heterogeneity landscape of malignant cells, we identified a three-subtype classification in HCC. Among them, S100A6+ tumor cells play a crucial role in metastasis. Targeting the feedback loop between tumor cells and fibroblasts is a promising anti-metastatic strategy.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Analyse sur cellule unique , Microenvironnement tumoral , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Humains , Régulation de l'expression des gènes tumoraux , Transition épithélio-mésenchymateuse/génétique , Animaux , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Hétérogénéité génétique , Souris , Lignée cellulaire tumorale , Pronostic , Analyse de profil d'expression de gènes , Transcriptome , Biologie informatique/méthodes , Métastase tumorale
2.
J Transl Med ; 22(1): 715, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090667

RÉSUMÉ

BACKGROUND: Synovial fibrosis is a common complication of knee osteoarthritis (KOA), a pathological process characterized by myofibroblast activation and excessive extracellular matrix (ECM) deposition. Fibroblast-like synoviocytes (FLSs) are implicated in KOA pathogenesis, contributing to synovial fibrosis through diverse mechanisms. Nuclear protein 1 (NUPR1) is a recently identified transcription factor with crucial roles in various fibrotic diseases. However, its molecular determinants in KOA synovial fibrosis remain unknown. This study aims to investigate the role of NUPR1 in KOA synovial fibrosis through in vivo and in vitro experiments. METHODS: We examined NUPR1 expression in the murine synovium and determined the impact of NUPR1 on synovial fibrosis by knockdown models in the destabilization of the medial meniscus (DMM)-induced KOA mouse model. TGF-ß was employed to induce fibrotic response and myofibroblast activation in mouse FLSs, and the role and molecular mechanisms in synovial fibrosis were evaluated under conditions of NUPR1 downexpression. Additionally, the pharmacological effect of NUPR1 inhibitor in synovial fibrosis was assessed using a surgically induced mouse KOA model. RESULTS: We found that NUPR1 expression increased in the murine synovium after DMM surgical operation. The adeno-associated virus (AAV)-NUPR1 shRNA promoted NUPR1 deficiency, attenuating synovial fibrosis, inhibiting synovial hyperplasia, and significantly reducing the expression of pro-fibrotic molecules. Moreover, the lentivirus-mediated NUPR1 deficiency alleviated synoviocyte proliferation and inhibited fibroblast to myofibroblast transition. It also decreased the expression of fibrosis markers α-SMA, COL1A1, CTGF, Vimentin and promoted the activation of the SMAD family member 3 (SMAD3) pathway. Importantly, trifluoperazine (TFP), a NUPR1 inhibitor, attenuated synovial fibrosis in DMM mice. CONCLUSIONS: These findings indicate that NUPR1 is an antifibrotic modulator in KOA, and its effect on anti-synovial fibrosis is partially mediated by SMAD3 signaling. This study reveals a promising target for developing novel antifibrotic treatment.


Sujet(s)
Fibroblastes , Fibrose , Transduction du signal , Protéine Smad-3 , Cellules synoviales , Animaux , Protéine Smad-3/métabolisme , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Souris de lignée C57BL , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Mâle , Gonarthrose/anatomopathologie , Gonarthrose/métabolisme , Modèles animaux de maladie humaine , Souris , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Protéines de liaison à l'ADN , Protéines tumorales
3.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 200-205, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097874

RÉSUMÉ

Keloids are defined as a benign dermal fibroproliferative disorder, with excessive fibroblast proliferation, and excessive overproduction of collagen. Although the heterogeneity during keloid development has been extensively studied, the heterogeneity across different skin states is still unclear. So, a global comparison across skin states is needed. In this study, we collected samples from 5 states of skin, including melanoma, cutaneous squamous cell carcinoma, keloid skin, scar skin, and healthy control samples. The heterogeneity of cell types and subtypes was analyzed and compared across 5 states, and we observed significant differences among them. Our results showed a cancer-like fibroblast, which is not in normal samples, may play an important role in antigen processing and presentation. We also noticed that the mesenchymal fibroblast increased in keloid samples, which highly expressed POSTN. And POSTN may participate in epithelial-mesenchymal transition and collagen overexpression to promote keloid growth. These findings help to understand the alteration among different skin states and provide potential genetic basis for keloid therapies.


Sujet(s)
Fibroblastes , Chéloïde , Tumeurs cutanées , Humains , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Chéloïde/génétique , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Tumeurs cutanées/génétique , Analyse sur cellule unique/méthodes , Peau/anatomopathologie , Peau/métabolisme , Molécules d'adhérence cellulaire/métabolisme , Molécules d'adhérence cellulaire/génétique , Transition épithélio-mésenchymateuse/génétique , Collagène/métabolisme , Mâle
4.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39125973

RÉSUMÉ

Altered ankyrin-R (AnkR; encoded by ANK1) expression is associated with diastolic function, left ventricular remodeling, and heart failure with preserved ejection fraction (HFpEF). First identified in erythrocytes, the role of AnkR in other tissues, particularly the heart, is less studied. Here, we identified the expression of both canonical and small isoforms of AnkR in the mouse myocardium. We demonstrate that cardiac myocytes primarily express small AnkR (sAnkR), whereas cardiac fibroblasts predominantly express canonical AnkR. As canonical AnkR expression in cardiac fibroblasts is unstudied, we focused on expression and localization in these cells. AnkR is expressed in both the perinuclear and cytoplasmic regions of fibroblasts with considerable overlap with the trans-Golgi network protein 38, TGN38, suggesting a potential role in trafficking. To study the role of AnkR in fibroblasts, we generated mice lacking AnkR in activated fibroblasts (Ank1-ifKO mice). Notably, Ank1-ifKO mice fibroblasts displayed reduced collagen compaction, supportive of a novel role of AnkR in normal fibroblast function. At the whole animal level, in response to a heart failure model, Ank1-ifKO mice displayed an increase in fibrosis and T-wave inversion compared with littermate controls, while preserving cardiac ejection fraction. Collagen type I fibers were decreased in the Ank1-ifKO mice, suggesting a novel function of AnkR in the maturation of collagen fibers. In summary, our findings illustrate the novel expression of AnkR in cardiac fibroblasts and a potential role in cardiac function in response to stress.


Sujet(s)
Ankyrines , Fibroblastes , Défaillance cardiaque , Souris knockout , Animaux , Défaillance cardiaque/métabolisme , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/génétique , Ankyrines/métabolisme , Ankyrines/génétique , Souris , Fibroblastes/métabolisme , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Mâle , Fibrose , Souris de lignée C57BL
5.
Gen Physiol Biophys ; 43(5): 469-484, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39140687

RÉSUMÉ

Ruthenium nitrosyl (Ru-NO) complexes are of interest as photoactive nitric oxide (NO) donor candidates for local therapeutic applications. NO plays a crucial regulatory role in skin homeostasis, concentration-dependently affecting processes like the proliferation, apoptosis, autophagy and redox balance. In this context, we investigated HE-10, a ruthenium-based photoinducible NO donor, for its pro-oxidant and cytotoxic effects under light and dark conditions in VH10 human foreskin fibroblast cells. We also tested its intracellular and extracellular NO-releasing function. Our study reveals a significant dose-dependent cytotoxic effect of HE-10, an increase in intracellular reactive oxygen and nitrogen species, and the occurrence of apoptosis in skin fibroblast cells. Furthermore, exposure to both increasing doses of HE-10 and white LED light led to substantial cellular events, including a significant induction of autophagy and G2/M phase cell cycle arrest. Paradoxically, these effects were not solely attributable to NO release based on DAF2-DA NO probe results, suggesting that intracellular photochemical reactions additional to NO photolysis contribute to HE-10's biological activity. This study shows that HE-10 exhibits both cytotoxic and potential therapeutic effects, depending on concentration and light exposure. These findings are crucial for developing targeted Ru-NO complex treatments for skin diseases and potentially certain types of skin cancer, where controlled NO release could be beneficial.


Sujet(s)
Fibroblastes , Monoxyde d'azote , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Monoxyde d'azote/métabolisme , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Ruthénium/composition chimique , Photosensibilisants/pharmacologie , Photosensibilisants/composition chimique , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Donneur d'oxyde nitrique/pharmacologie , Donneur d'oxyde nitrique/composition chimique , Relation dose-effet des médicaments , Lumière
6.
PLoS One ; 19(8): e0308723, 2024.
Article de Anglais | MEDLINE | ID: mdl-39133718

RÉSUMÉ

Fibrosis is a complex pathological process that can lead to the permanent loss of biological function, with P2ry2 playing a crucial role in this process. Long non-coding RNAs (lncRNAs) have been reported to play an critically important role in the fibrotic process. However, it remains unclear whether lncRNAs can regulate fibrosis through P2ry2. In this study, we detected the expression of the long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (lnc-MALAT1). We investigated the expression patterns of lnc-MALAT1 and P2ry2 in denervated skeletal muscle, a classical model of fibrosis. Additionally, we utilized a TGF-ß-mediated fibrosis model in NIH/3T3 cells to examine the effects of lnc-MALAT1 and P2ry2 on fibroblast activation and the underlying regulatory mechanisms in vitro. Our results demonstrated that the expression levels of lnc-MALAT1 and P2ry2 were consistently elevated in denervated skeletal muscle, correlating with the degree of fibrosis. In vitro experiments confirmed the regulatory effect of lnc-MALAT1 on P2ry2. Furthermore, we identified miR-335-3p as a potential key molecule in the regulatory relationship of lnc-MALAT1/P2ry2. Dual luciferase reporter assays and AGO2-RIP verified the molecular sponging effect of lnc-MALAT1 on miR-335-3p. Additionally, we validated the regulation of the lnc-MALAT1/miR-335-3p/P2ry2 axis through experimental approaches. In conclusion, our study identified a crucial role of lnc-MALAT1/miR-335-3p/P2ry2 axis in fibroblast activation, providing a promising treatment option against the fibrosis.


Sujet(s)
Fibroblastes , Fibrose , microARN , ARN long non codant , Animaux , ARN long non codant/génétique , ARN long non codant/métabolisme , microARN/génétique , microARN/métabolisme , Souris , Fibroblastes/métabolisme , Cellules NIH 3T3 , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Mâle , Souris de lignée C57BL , Facteur de croissance transformant bêta/métabolisme , Régulation de l'expression des gènes ,
7.
Cancer Cell ; 42(8): 1370-1385.e9, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39137726

RÉSUMÉ

Tertiary lymphoid structures (TLSs) are associated with enhanced immunity in tumors. However, their formation and functions in colorectal cancer liver metastasis (CRLM) remain unclear. Here, we reveal that intra- and peri-tumor mature TLSs (TLS+) are associated with improved clinical outcomes than TLS- tumors. Using single-cell-RNA-sequencing and spatial-enhanced-resolution-omics-sequencing (Stereo-seq), we reveal that TLS+ tumors are enriched with IgG+ plasma cells (PCs), while TLS- tumors are characterized with IgA+ PCs. By generating TLS-associated PC-derived monoclonal antibodies in vitro, we show that TLS-PCs secrete tumor-targeting antibodies. As the proof-of-concept, we demonstrate the anti-tumor activities of TLS-PC-mAb6 antibody in humanized mouse model of colorectal cancer. We identify a fibroblast lineage secreting CCL19 that facilitates lymphocyte trafficking to TLSs. CCL19 treatment promotes TLS neogenesis and prevents tumor growth in mice. Our data uncover the central role of CCL19+ fibroblasts in TLS formation, which in turn generates therapeutic antibodies to restrict CRLM.


Sujet(s)
Chimiokine CCL19 , Tumeurs colorectales , Immunoglobuline G , Tumeurs du foie , Structures lymphoïdes tertiaires , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/immunologie , Tumeurs colorectales/métabolisme , Animaux , Structures lymphoïdes tertiaires/immunologie , Structures lymphoïdes tertiaires/anatomopathologie , Humains , Tumeurs du foie/immunologie , Tumeurs du foie/secondaire , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Souris , Immunoglobuline G/immunologie , Chimiokine CCL19/métabolisme , Chimiokine CCL19/génétique , Fibroblastes/métabolisme , Fibroblastes/immunologie , Anticorps monoclonaux/pharmacologie , Plasmocytes/immunologie , Plasmocytes/métabolisme , Femelle , Lignée cellulaire tumorale
8.
Int J Mol Sci ; 25(15)2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39126066

RÉSUMÉ

Pathogenic variations in the fused in sarcoma (FUS) gene are associated with rare and aggressive forms of amyotrophic lateral sclerosis (ALS). As FUS-ALS is a dominant disease, a targeted, allele-selective approach to FUS knockdown is most suitable. Antisense oligonucleotides (AOs) are a promising therapeutic platform for treating such diseases. In this study, we have explored the potential for allele-selective knockdown of FUS. Gapmer-type AOs targeted to two common neutral polymorphisms in FUS were designed and evaluated in human fibroblasts. AOs had either methoxyethyl (MOE) or thiomorpholino (TMO) modifications. We found that the TMO modification improved allele selectivity and efficacy for the lead sequences when compared to the MOE counterparts. After TMO-modified gapmer knockdown of the target allele, up to 93% of FUS transcripts detected were from the non-target allele. Compared to MOE-modified AOs, the TMO-modified AOs also demonstrated reduced formation of structured nuclear inclusions and SFPQ aggregation that can be triggered by phosphorothioate-containing AOs. How overall length and gap length of the TMO-modified AOs affected allele selectivity, efficiency and off-target gene knockdown was also evaluated. We have shown that allele-selective knockdown of FUS may be a viable therapeutic strategy for treating FUS-ALS and demonstrated the benefits of the TMO modification for allele-selective applications.


Sujet(s)
Allèles , Sclérose latérale amyotrophique , Oligonucléotides antisens , Protéine FUS de liaison à l'ARN , Humains , Oligonucléotides antisens/usage thérapeutique , Oligonucléotides antisens/génétique , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/traitement médicamenteux , Sclérose latérale amyotrophique/thérapie , Protéine FUS de liaison à l'ARN/génétique , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Techniques de knock-down de gènes , Morpholinos/usage thérapeutique , Morpholinos/génétique
9.
Mol Med Rep ; 30(4)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39129301

RÉSUMÉ

Chitinase­3 like­protein­1 (CHI3L1), a glycoprotein belonging to the glycoside hydrolase family 18, binds to chitin; however, this protein lacks chitinase activity. Although CHI3L1 is not an enzyme capable of degrading chitin, it plays significant roles in abnormal glucose and lipid metabolism, indicating its involvement in metabolic disorders. In addition, CHI3L1 is considered a key player in inflammatory diseases, with clinical data suggesting its potential as a predictor of cardiovascular disease. CHI3L1 regulates the inflammatory response of various cell types, including macrophages, vascular smooth muscle cells and fibroblasts. In addition, CHI3L1 participates in vascular remodeling and fibrosis, contributing to the pathogenesis of cardiovascular disease. At present, research is focused on elucidating the role of CHI3L1 in cardiovascular disease. The present systematic review was conducted to comprehensively evaluate the effects of CHI3L1 on cardiovascular cells, and determine the potential implications in the occurrence and progression of cardiovascular disease. The present study may further the understanding of the involvement of CHI3L1 in cardiovascular pathology, demonstrating its potential as a therapeutic target or biomarker in the management of cardiovascular disease.


Sujet(s)
Marqueurs biologiques , Maladies cardiovasculaires , Protéine-1 similaire à la chitinase-3 , Humains , Maladies cardiovasculaires/métabolisme , Maladies cardiovasculaires/anatomopathologie , Protéine-1 similaire à la chitinase-3/métabolisme , Animaux , Fibroblastes/métabolisme
10.
Nat Commun ; 15(1): 6703, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39112444

RÉSUMÉ

Extreme myopia (EM), defined as a spherical equivalent (SE) ≤ -10.00 diopters (D), is one of the leading causes of sight impairment. Known EM-associated variants only explain limited risk and are inadequate for clinical decision-making. To discover risk genes, we performed a whole-exome sequencing (WES) on 449 EM individuals and 9606 controls. We find a significant excess of rare protein-truncating variants (PTVs) in EM cases, enriched in the retrograde vesicle-mediated transport pathway. Employing single-cell RNA-sequencing (scRNA-seq) and a single-cell polygenic burden score (scPBS), we pinpointed PI16 + /SFRP4+ fibroblasts as the most relevant cell type. We observed that KDELR3 is highly expressed in scleral fibroblast and involved in scleral extracellular matrix (ECM) organization. The zebrafish model revealed that kdelr3 downregulation leads to elongated ocular axial length and increased lens diameter. Together, our study provides insight into the genetics of EM in humans and highlights KDELR3's role in EM pathogenesis.


Sujet(s)
, Mutation , Danio zébré , Humains , Animaux , Danio zébré/génétique , Mâle , Femelle , Fibroblastes/métabolisme , Exome/génétique , Étude d'association pangénomique , Adulte , Myopie/génétique , Myopie/métabolisme , Myopie/anatomopathologie , Sclère/métabolisme , Sclère/anatomopathologie , Matrice extracellulaire/métabolisme , Matrice extracellulaire/génétique , Prédisposition génétique à une maladie , Analyse sur cellule unique , Études cas-témoins , Enfant , Jeune adulte
11.
Cell Death Dis ; 15(8): 584, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122678

RÉSUMÉ

TNF is a potent cytokine known for its involvement in physiology and pathology. In Rheumatoid Arthritis (RA), persistent TNF signals cause aberrant activation of synovial fibroblasts (SFs), the resident cells crucially involved in the inflammatory and destructive responses of the affected synovial membrane. However, the molecular switches that control the pathogenic activation of SFs remain poorly defined. Cyld is a major component of deubiquitination (DUB) machinery regulating the signaling responses towards survival/inflammation and programmed necrosis that induced by cytokines, growth factors and microbial products. Herein, we follow functional genetic approaches to understand how Cyld affects arthritogenic TNF signaling in SFs. We demonstrate that in spontaneous and induced RA models, SF-Cyld DUB deficiency deteriorates arthritic phenotypes due to increased levels of chemokines, adhesion receptors and bone-degrading enzymes generated by mutant SFs. Mechanistically, Cyld serves to restrict the TNF-induced hyperactivation of SFs by limiting Tak1-mediated signaling, and, therefore, leading to supervised NF-κB and JNK activity. However, Cyld is not critically involved in the regulation of TNF-induced death of SFs. Our results identify SF-Cyld as a regulator of TNF-mediated arthritis and inform the signaling landscape underpinning the SF responses.


Sujet(s)
Polyarthrite rhumatoïde , Deubiquitinating enzyme CYLD , Fibroblastes , I-kappa B Kinase , MAP Kinase Kinase Kinases , Transduction du signal , Membrane synoviale , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Deubiquitinating enzyme CYLD/métabolisme , Deubiquitinating enzyme CYLD/génétique , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Animaux , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Souris , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Humains , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée C57BL , Souris knockout
12.
Nat Commun ; 15(1): 6820, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122702

RÉSUMÉ

Biomaterial wound dressings, such as hydrogels, interact with host cells to regulate tissue repair. This study investigates how crosslinking of gelatin-based hydrogels influences immune and stromal cell behavior and wound healing in female mice. We observe that softer, lightly crosslinked hydrogels promote greater cellular infiltration and result in smaller scars compared to stiffer, heavily crosslinked hydrogels. Using single-cell RNA sequencing, we further show that heavily crosslinked hydrogels increase inflammation and lead to the formation of a distinct macrophage subpopulation exhibiting signs of oxidative activity and cell fusion. Conversely, lightly crosslinked hydrogels are more readily taken up by macrophages and integrated within the tissue. The physical properties differentially affect macrophage and fibroblast interactions, with heavily crosslinked hydrogels promoting pro-fibrotic fibroblast activity that drives macrophage fusion through RANKL signaling. These findings suggest that tuning the physical properties of hydrogels can guide cellular responses and improve healing, offering insights for designing better biomaterials for wound treatment.


Sujet(s)
Fibroblastes , Hydrogels , Macrophages , Cicatrisation de plaie , Animaux , Hydrogels/composition chimique , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Souris , Femelle , Communication cellulaire/effets des médicaments et des substances chimiques , Matériaux biocompatibles/composition chimique , Ligand de RANK/métabolisme , Souris de lignée C57BL , Réactifs réticulants/composition chimique , Gélatine/composition chimique , Inflammation/métabolisme , Inflammation/anatomopathologie
13.
Proc Natl Acad Sci U S A ; 121(34): e2320257121, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39150784

RÉSUMÉ

Lysosomal degradation pathways coordinate the clearance of superfluous and damaged cellular components. Compromised lysosomal degradation is a hallmark of many degenerative diseases, including lysosomal storage diseases (LSDs), which are caused by loss-of-function mutations within both alleles of a lysosomal hydrolase, leading to lysosomal substrate accumulation. Gaucher's disease, characterized by <15% of normal glucocerebrosidase function, is the most common LSD and is a prominent risk factor for developing Parkinson's disease. Here, we show that either of two structurally distinct small molecules that modulate PIKfyve activity, identified in a high-throughput cellular lipid droplet clearance screen, can improve glucocerebrosidase function in Gaucher patient-derived fibroblasts through an MiT/TFE transcription factor that promotes lysosomal gene translation. An integrated stress response (ISR) antagonist used in combination with a PIKfyve modulator further improves cellular glucocerebrosidase activity, likely because ISR signaling appears to also be slightly activated by treatment by either small molecule at the higher doses employed. This strategy of combining a PIKfyve modulator with an ISR inhibitor improves mutant lysosomal hydrolase function in cellular models of additional LSD.


Sujet(s)
Fibroblastes , Glucosylceramidase , Maladies lysosomiales , Lysosomes , Phosphatidylinositol 3-kinases , Humains , Phosphatidylinositol 3-kinases/métabolisme , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Glucosylceramidase/métabolisme , Glucosylceramidase/génétique , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Maladies lysosomiales/traitement médicamenteux , Maladies lysosomiales/génétique , Maladies lysosomiales/métabolisme , Maladie de Gaucher/traitement médicamenteux , Maladie de Gaucher/génétique , Maladie de Gaucher/métabolisme , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie
14.
Sci Rep ; 14(1): 18939, 2024 08 15.
Article de Anglais | MEDLINE | ID: mdl-39147768

RÉSUMÉ

Rheumatoid arthritis (RA) and arthrofibrosis (AF) are both chronic synovial hyperplasia diseases that result in joint stiffness and contractures. They shared similar symptoms and many common features in pathogenesis. Our study aims to perform a comprehensive analysis between RA and AF and identify novel drugs for clinical use. Based on the text mining approaches, we performed a correlation analysis of 12 common joint diseases including arthrofibrosis, gouty arthritis, infectious arthritis, juvenile idiopathic arthritis, osteoarthritis, post infectious arthropathies, post traumatic osteoarthritis, psoriatic arthritis, reactive arthritis, rheumatoid arthritis, septic arthritis, and transient arthritis. 5 bulk sequencing datasets and 4 single-cell sequencing datasets of RA and AF were integrated and analyzed. A novel drug repositioning method was found for drug screening, and text mining approaches were used to verify the identified drugs. RA and AF performed the highest gene similarity (0.77) and functional ontology similarity (0.84) among all 12 joint diseases. We figured out that they share the same key pathogenic cell including CD34 + sublining fibroblasts (CD34-SLF) and DKK3 + sublining fibroblasts (DKK3-SLF). Potential therapeutic target database (PTTD) was established with the differential expressed genes (DEGs) of these key pathogenic cells. Based on the PTTD, 15 potential drugs for AF and 16 potential drugs for RA were identified. This work provides a new perspective on AF and RA study which enhances our understanding of their pathogenesis. It also shed light on their underlying mechanism and open new avenues for drug repositioning studies.


Sujet(s)
Polyarthrite rhumatoïde , Fibrose , Membrane synoviale , Humains , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Repositionnement des médicaments , Microenvironnement cellulaire/effets des médicaments et des substances chimiques , Fouille de données
15.
Sci Rep ; 14(1): 18345, 2024 08 07.
Article de Anglais | MEDLINE | ID: mdl-39112598

RÉSUMÉ

Pressure ulcers (PU) are caused by persistent long-term pressure, which compromises the integrity of the epidermis, dermis, and subcutaneous adipose tissue layer by layer, making it difficult to heal. Platelet products such as platelet lysate (PL) can promote tissue regeneration by secreting numerous growth factors based on clinical studies on skin wound healing. However, the components of PL are difficult to retain in wounds. Gelatin methacrylate (GelMA) is a photopolymerizable hydrogel that has lately emerged as a promising material for tissue engineering and regenerative medicine. The PL liquid was extracted, flow cytometrically detected for CD41a markers, and evenly dispersed in the GelMA hydrogel to produce a surplus growth factor hydrogel system (PL@GM). The microstructure of the hydrogel system was observed under a scanning electron microscope, and its sustained release efficiency and biological safety were tested in vitro. Cell viability and migration of human dermal fibroblasts, and tube formation assays of human umbilical vein endothelial cells were applied to evaluate the ability of PL to promote wound healing and regeneration in vitro. Real-time polymerase chain reaction (PCR) and western blot analyses were performed to elucidate the skin regeneration mechanism of PL. We verified PL's therapeutic effectiveness and histological analysis on the PU model. PL promoted cell viability, migration, wound healing and angiogenesis in vitro. Real-time PCR and western blot indicated PL suppressed inflammation and promoted collagen I synthesis by activating STAT3. PL@GM hydrogel system demonstrated optimal biocompatibility and favorable effects on essential cells for wound healing. PL@GM also significantly stimulated PU healing, skin regeneration, and the formation of subcutaneous collagen and blood vessels. PL@GM could accelerate PU healing by promoting fibroblasts to migrate and secrete collagen and endothelial cells to vascularize. PL@GM promises to be an effective and convenient treatment modality for PU, like chronic wound treatment.


Sujet(s)
, Plaquettes , Gélatine , Méthacrylates , Escarre , Peau , Cicatrisation de plaie , Animaux , Humains , Souris , /effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Gélatine/composition chimique , Gélatine/pharmacologie , Cellules endothéliales de la veine ombilicale humaine , Hydrogels/composition chimique , Méthacrylates/composition chimique , Méthacrylates/pharmacologie , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Escarre/thérapie , Régénération/effets des médicaments et des substances chimiques , Peau/vascularisation , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Peau/anatomopathologie , Facteur de transcription STAT-3/métabolisme , Cicatrisation de plaie/effets des médicaments et des substances chimiques
16.
Cells ; 13(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39120290

RÉSUMÉ

Mutations in human CILK1 (ciliogenesis associated kinase 1) are linked to ciliopathies and epilepsy. Homozygous point and nonsense mutations that extinguish kinase activity impair primary cilia function, whereas mutations outside the kinase domain are not well understood. Here, we produced a knock-in mouse equivalent to the human CILK1 A615T variant identified in juvenile myoclonic epilepsy (JME). This residue is in the intrinsically disordered C-terminal region of CILK1 separate from the kinase domain. Mouse embryo fibroblasts (MEFs) with either heterozygous or homozygous A612T mutant alleles exhibited a higher ciliation rate, shorter individual cilia, and upregulation of ciliary Hedgehog signaling. Thus, a single A612T mutant allele was sufficient to impair primary cilia and ciliary signaling in MEFs. Gene expression profiles of wild-type versus mutant MEFs revealed profound changes in cilia-related molecular functions and biological processes. The CILK1 A615T mutant protein was not increased to the same level as the wild-type protein when co-expressed with scaffold protein KATNIP (katanin-interacting protein). Our data show that KATNIP regulation of a JME-associated single-residue variant of CILK1 is compromised, and this impairs the maintenance of primary cilia and Hedgehog signaling.


Sujet(s)
Cils vibratiles , Épilepsie , Protéines Hedgehog , Transduction du signal , Cils vibratiles/métabolisme , Animaux , Protéines Hedgehog/métabolisme , Souris , Épilepsie/génétique , Épilepsie/métabolisme , Épilepsie/anatomopathologie , Humains , Fibroblastes/métabolisme , Mutation/génétique , Protein-Serine-Threonine Kinases
17.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39125660

RÉSUMÉ

Colostrum is gaining popularity in cosmetic products. The present study compared the composition and selected biological properties of colostrum from Polish sheep (colostrum 1) and Swiss sheep (colostrum 2), particularly those that can affect healthy or diseased skin. The antioxidant activity of the colostrums was measured using ABTS and DPPH assays. The effect on the proliferation of human skin fibroblasts, neonatal epidermal keratinocytes, and human diabetic fibroblast (dHF) cells isolated from diabetic foot ulcers was also assayed in vitro by MTT and Presto Blue tests, respectively. The colostrum simulated dHF cell proliferation by up to 115.4%. The highest used concentration of colostrum 1 stimulated normal fibroblast proliferation by 191.2% (24 h) and 222.2% (48 h). Both colostrums inhibited epidermal keratinocyte viability. The influence of the colostrums on the expression of genes related to proliferation (Ki67) and immune response (IL-6, PTGS-2, TSG-6) in dHF cells were compared. Colostrum 1 increased the rate of wound closure (scar test). Analysis of total fat, protein and fatty acid content found the Polish colostrum to be a richer source of fat than the Swiss colostrum, which contained a larger amount of protein. Both colostrums exhibit properties that suggest they could be effective components in cosmetic or medicinal formulations for skin care, especially supporting its regeneration, rejuvenation, and wound healing.


Sujet(s)
Prolifération cellulaire , Colostrum , Fibroblastes , Kératinocytes , Hygiène de la peau , Colostrum/composition chimique , Animaux , Ovis , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Hygiène de la peau/méthodes , Antioxydants/pharmacologie , Femelle , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Peau/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Grossesse , Administration par voie topique , Pied diabétique/thérapie , Pied diabétique/traitement médicamenteux , Pied diabétique/métabolisme , Cellules cultivées
18.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39125691

RÉSUMÉ

Cell immortalization, a hallmark of cancer development, is a process that cells can undergo on their path to carcinogenesis. Spontaneously immortalized mouse embryonic fibroblasts (MEFs) have been used for decades; however, changes in the global transcriptome during this process have been poorly described. In our research, we characterized the poly-A RNA transcriptome changes after spontaneous immortalization. To this end, differentially expressed genes (DEGs) were screened using DESeq2 and characterized by gene ontology enrichment analysis and protein-protein interaction (PPI) network analysis to identify the potential hub genes. In our study, we identified changes in the expression of genes involved in proliferation regulation, cell adhesion, immune response and transcriptional regulation in immortalized MEFs. In addition, we performed a comparative analysis with previously reported MEF immortalization data, where we propose a predicted gene regulatory network model in immortalized MEFs based on the altered expression of Mapk11, Cdh1, Chl1, Zic1, Hoxd10 and the novel hub genes Il6 and Itgb2.


Sujet(s)
Fibroblastes , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Transcriptome , Animaux , Souris , Fibroblastes/métabolisme , Cartes d'interactions protéiques/génétique , Embryon de mammifère/métabolisme , Gene Ontology
19.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39125720

RÉSUMÉ

Freesia refracta (FR), a perennial flower of the Iris family (Iridaceae), is widely used in cosmetics despite limited scientific evidence of its skin benefits and chemical composition, particularly of FR callus extract (FCE). This study identified biologically active compounds in FCE and assessed their skin benefits, focusing on anti-aging. FR calli were cultured, extracted with water at 40 °C, and analyzed using Centrifugal Partition Chromatography (CPC), Nuclear Magnetic Resonance (NMR), and HCA, revealing key compounds, namely nicotinamide and pyroglutamic acid. FCE significantly increased collagen I production by 52% in normal and aged fibroblasts and enhanced fibroblast-collagen interaction by 37%. An in vivo study of 43 female volunteers demonstrated an 11.1% reduction in skin roughness and a 2.3-fold increase in collagen density after 28 days of cream application containing 3% FCE. Additionally, the preservation tests of cosmetics containing FCE confirmed their stability over 12 weeks. These results suggest that FCE offers substantial anti-aging benefits by enhancing collagen production and fibroblast-collagen interactions. These findings highlighted the potential of FCE in cosmetic applications, providing significant improvements in skin smoothness and overall appearance. This study fills a gap in the scientific literature regarding the skin benefits and chemical composition of FR callus extract, supporting its use in the development of effective cosmeceuticals.


Sujet(s)
Fibroblastes , Stress oxydatif , Extraits de plantes , Vieillissement de la peau , Peau , Vieillissement de la peau/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Humains , Femelle , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Adulte , Collagène/métabolisme , Cosmétiques/pharmacologie , Adulte d'âge moyen , Nicotinamide/pharmacologie , Acide pidolique/analogues et dérivés , Acide pidolique/pharmacologie , Acide pidolique/métabolisme
20.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39125782

RÉSUMÉ

Ferroptosis is a form of iron-dependent regulated cell death caused by the accumulation of lipid peroxides. In this review, we summarize research on the impact of ferroptosis on disease models and isolated cells in various types of arthritis. While most studies have focused on rheumatoid arthritis (RA) and osteoarthritis (OA), there is limited research on spondylarthritis and crystal arthropathies. The effects of inducing or inhibiting ferroptosis on the disease strongly depend on the studied cell type. In the search for new therapeutic targets, inhibiting ferroptosis in chondrocytes might have promising effects for any type of arthritis. On the other hand, ferroptosis induction may also lead to a desired decrease of synovial fibroblasts in RA. Thus, ferroptosis research must consider the cell-type-specific effects on arthritis. Further investigation is needed to clarify these complexities.


Sujet(s)
Ferroptose , Arthrose , Humains , Animaux , Arthrose/métabolisme , Arthrose/anatomopathologie , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Arthrite/métabolisme , Arthrite/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fer/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE