Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 3 de 3
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Stem Cell Res Ther ; 12(1): 586, 2021 11 24.
Article de Anglais | MEDLINE | ID: mdl-34819138

RÉSUMÉ

BACKGROUND: Injection of autologous adipose tissue (AT) has recently been demonstrated to be an effective and safe treatment for anal fistulas. AT mesenchymal stem cells (AT-MSCs) mediate the healing process, but the relationship between molecular characteristics of AT-MSCs of the injected AT and fistula healing has not been adequately studied. Thus we aimed to characterize the molecular and functional properties of AT-MSCs isolated from autologous AT injected as a treatment of cryptogenic high transsphincteric perianal fistulas and correlate these findings to the healing process. METHODS: 27 patients (age 45 ± 2 years) diagnosed with perianal fistula were enrolled in the study and treated with autologous AT injected around the anal fistula tract. AT-MSCs were isolated for cellular and molecular analyses. The fistula healing was evaluated by MRI scanning after 6 months of treatment. AT-MSC phenotype was compared between responders and non-responders with respect to fistula healing. RESULTS: 52% of all patients exhibited clinical healing of the fistulas as evaluated 6 months after last injection. Cultured AT-MSCs in the responder group had a lower short-term proliferation rate and higher osteoblast differentiation potential compared to non-responder AT-MSCs. On the other hand, adipocyte differentiation potential of AT-MSCs was higher in non-responder group. Interestingly, AT-MSCs of responders exhibited lower expression of inflammatory and senescence associated genes such as IL1B, NFKB, CDKN2A, TPB3,TGFB1. CONCLUSION: Our data suggest that cellular quality of the injected AT-MSCs including cell proliferation, differentiation capacity and secretion of proinflammatory molecules may provide a possible mechanism underlying fistula healing. Furthermore, these biomarkers may be useful to predict a positive fistula healing outcome. TRIAL REGISTRATION: NTC04834609, Registered 6 April 2021. https://clinicaltrials.gov/ct2/show/NCT04834609.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Fistule rectale , Tissu adipeux , Adulte , Humains , Transplantation de cellules souches mésenchymateuses/méthodes , Adulte d'âge moyen , Fistule rectale/génétique , Fistule rectale/thérapie , Résultat thérapeutique
2.
Cell Rep ; 32(2): 107877, 2020 07 14.
Article de Anglais | MEDLINE | ID: mdl-32668244

RÉSUMÉ

Evolutionarily conserved SCAN (named after SRE-ZBP, CTfin51, AW-1, and Number 18 cDNA)-domain-containing zinc finger transcription factors (ZSCAN) have been found in both mouse and human genomes. Zscan4 is transiently expressed during zygotic genome activation (ZGA) in preimplantation embryos and induced pluripotent stem cell (iPSC) reprogramming. However, little is known about the mechanism of Zscan4 underlying these processes of cell fate control. Here, we show that Zscan4f, a representative of ZSCAN proteins, is able to recruit Tet2 through its SCAN domain. The Zscan4f-Tet2 interaction promotes DNA demethylation and regulates the expression of target genes, particularly those encoding glycolytic enzymes and proteasome subunits. Zscan4f regulates metabolic rewiring, enhances proteasome function, and ultimately promotes iPSC generation. These results identify Zscan4f as an important partner of Tet2 in regulating target genes and promoting iPSC generation and suggest a possible and common mechanism shared by SCAN family transcription factors to recruit ten-eleven translocation (TET) DNA dioxygenases to regulate diverse cellular processes, including reprogramming.


Sujet(s)
Reprogrammation cellulaire/génétique , Protéines de liaison à l'ADN/métabolisme , Homéostasie protéique/génétique , Protéines proto-oncogènes/métabolisme , Facteurs de transcription/métabolisme , Transcription génétique , Animaux , Séquence nucléotidique , ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Dioxygenases , Glycolyse/génétique , Cellules HEK293 , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules MCF-7 , Souris de lignée C57BL , Proteasome endopeptidase complex/métabolisme , Liaison aux protéines , Domaines protéiques , Protéines proto-oncogènes/génétique , Régulation positive
3.
FASEB J ; 33(7): 8321-8334, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-30970214

RÉSUMÉ

Although the mouse strain Murphy Roths Large (MRL/MpJ) possesses high regenerative potential, the mechanism of tissue regeneration, including skeletal muscle, in MRL/MpJ mice after injury is still unclear. Our previous studies have shown that muscle-derived stem/progenitor cell (MDSPC) function is significantly enhanced in MRL/MpJ mice when compared with MDSPCs isolated from age-matched wild-type (WT) mice. Using mass spectrometry-based proteomic analysis, we identified increased expression of hypoxia-inducible factor (HIF) 1α target genes (expression of glycolytic factors and antioxidants) in sera from MRL/MpJ mice compared with WT mice. Therefore, we hypothesized that HIF-1α promotes the high muscle healing capacity of MRL/MpJ mice by increasing the potency of MDSPCs. We demonstrated that treating MRL/MpJ MDSPCs with dimethyloxalylglycine and CoCl2 increased the expression of HIF-1α and target genes, including angiogenic and cell survival genes. We also observed that HIF-1α activated the expression of paired box (Pax)7 through direct interaction with the Pax7 promoter. Furthermore, we also observed a higher myogenic potential of MDSPCs derived from prolyl hydroxylase (Phd) 3-knockout (Phd3-/-) mice, which displayed higher stability of HIF-1α. Taken together, our findings suggest that HIF-1α is a major determinant in the increased MDSPC function of MRL/MpJ mice through enhancement of cell survival, proliferation, and myogenic differentiation.-Sinha, K. M., Tseng, C., Guo, P., Lu, A., Pan, H., Gao, X., Andrews, R., Eltzschig, H., Huard, J. Hypoxia-inducible factor 1α (HIF-1α) is a major determinant in the enhanced function of muscle-derived progenitors from MRL/MpJ mice.


Sujet(s)
Régulation de l'expression des gènes , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Muscles squelettiques/physiologie , Régénération , Cellules souches/métabolisme , Animaux , Cobalt/pharmacologie , Glycine/analogues et dérivés , Glycine/pharmacologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Souris , Souris knockout , Muscles squelettiques/anatomopathologie , Facteur de transcription PAX7/génétique , Facteur de transcription PAX7/métabolisme , Procollagen-Proline Dioxygenase/génétique , Procollagen-Proline Dioxygenase/métabolisme , Cellules souches/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...