Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Stem Cell Res ; 79: 103476, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38941882

RÉSUMÉ

Cardiovascular disease remains a global health concern. Stem cell therapy utilizing human cardiac progenitor cells (hCPCs) shows promise in treating cardiac vascular disease. However, limited availability and senescence of hCPCs hinder their widespread use. To address these challenges, researchers are exploring innovative approaches. In this study, a bioengineered cell culture plate was developed to mimic the natural cardiac tissue microenvironment. It was coated with a combination of extracellular matrix (ECM) peptide motifs and mussel adhesive protein (MAP). The selected ECM peptide motifs, derived from fibronectin and vitronectin, play crucial roles in hCPCs. Results revealed that the Fibro-P and Vitro-P coated plates significantly improved hCPC adhesion, proliferation, migration, and differentiation compared to uncoated plates. Additionally, long-term culture on the coated plates delayed cellular senescence and maintained hCPC stemness. These enhancements were attributed to the activation of integrin downstream signaling pathways. The findings suggest that the engineered ECM peptide motif-MAP-coated plates hold potential for enhancing the therapeutic efficacy of stem cell-based therapies in cardiac tissue engineering and regenerative medicine.


Sujet(s)
Fibronectines , Cellules souches , Vitronectine , Vitronectine/métabolisme , Humains , Fibronectines/métabolisme , Cellules souches/cytologie , Cellules souches/métabolisme , Différenciation cellulaire , Techniques de culture cellulaire/méthodes , Cellules cultivées , Prolifération cellulaire , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Adhérence cellulaire , Peptides
3.
Biosens Bioelectron ; 246: 115838, 2024 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-38042052

RÉSUMÉ

Stem cell technology holds immense potential for revolutionizing medicine, particularly in regenerative treatment for heart disease. The unique capacity of stem cells to differentiate into diverse cell types offers promise in repairing damaged tissues and implanting organs. Ensuring the quality of differentiated cells, essential for specific functions, demands in-depth analysis. However, this process consumes time and incurs substantial costs while invasive methods may alter stem cell features during differentiation and deplete cell numbers. To address these challenges, we propose a non-invasive strategy, using cellular respiration, to assess the quality of differentiation-induced stem cells, notably cardiovascular stem cells. This evaluation employs an electronic nose (E-Nose) and neural pattern separation (NPS). Our goal is to assess differentiation-induced cardiac stem cells (DICs) quality through E-Nose data analysis and compare it with standard commercial human cells (SCHCs). Sensitivity and specificity were evaluated by interacting SCHCs and DICs with the E-Nose, achieving over 90% classification accuracy. Employing selective combinations optimized by NPS, E-Nose successfully classified all six cell types. Consequently, the relative similarity among DICs like cardiomyocytes, endothelial cells with SCHCs was established relied on comparing response data from the E-Nose sensor without resorting to complex evaluations.


Sujet(s)
Techniques de biocapteur , Nez électronique , Humains , Cellules endothéliales , Différenciation cellulaire , Cellules souches
5.
Oral Dis ; 2023 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-37724481

RÉSUMÉ

OBJECTIVE: This study investigated the effects of Lactobacillus fermentum BELF11 on periodontitis in mice (LIP). METHODS: Sixty mice were randomly assigned to a control group (CTL), LIP/PBS group (LIP and PBS applied), or LIP/BELF11 group (LIP and L. fermentum BELF11 applied). For 14 days, PBS or L. fermentum BELF11 was applied twice daily to the mice in the LIP/PBS or LIP/BELF11 group, respectively. After 14 days, radiographic, histological, and pro-inflammatory cytokine assessments were conducted. RESULTS: The LIP/PBS and LIP/BELF11 groups demonstrated greater alveolar bone loss than the CTL group (p < 0.05). The LIP/BELF11 group showed significantly reduced alveolar bone loss on the mesial side compared to the LIP/PBS group. Histologically, the LIP/BELF11 group showed consistent patterns of connective tissue fiber arrangement, lower levels of inflammatory infiltration, less alveolar bone loss, and higher alveolar bone density than the LIP/PBS group, despite showing more signs of destruction than the CTL group. The LIP/BELF11 group also exhibited significantly lower levels of pro-inflammatory cytokines than the LIP/PBS group. CONCLUSIONS: L. fermentum BELF11 inhibits alveolar bone loss and periodontitis progression by regulating pro-inflammatory cytokine production. These findings suggest that L. fermentum BELF11 may be a potential adjunctive therapy in periodontal treatment.

6.
Cells ; 12(15)2023 08 05.
Article de Anglais | MEDLINE | ID: mdl-37566085

RÉSUMÉ

Endothelial progenitor cell (EPC)-based stem cell therapy is a promising therapeutic strategy for vascular diseases. However, continuous in vitro expansion for clinical studies induces the loss of EPC functionality due to aging. In this study, we investigated the effects of StemRegenin-1 (SR-1), an antagonist of aryl hydrocarbon receptor (AhR), on replicative senescence in EPCs. We found that SR-1 maintained the expression of EPC surface markers, including stem cell markers, such as CD34, c-Kit, and CXCR4. Moreover, SR-1 long-term-treated EPCs preserved their characteristics. Subsequently, we demonstrated that SR-1 showed that aging phenotypes were reduced through senescence-associated phenotypes, such as ß-galactosidase activity, SMP30, p21, p53, and senescence-associated secretory phenotype (SASP). SR-1 treatment also increased the proliferation, migration, and tube-forming capacity of senescent EPCs. SR-1 inhibited the AhR-mediated cytochrome P450 (CYP)1A1 expression, reactive-oxygen species (ROS) production, and DNA damage under oxidative stress conditions in EPCs. Furthermore, as a result of CYP1A1-induced ROS inhibition, it was found that accumulated intracellular ROS were decreased in senescent EPCs. Finally, an in vivo Matrigel plug assay demonstrated drastically enhanced blood vessel formation via SR-1-treated EPCs. In summary, our results suggest that SR-1 contributes to the protection of EPCs against cellular senescence.


Sujet(s)
Progéniteurs endothéliaux , Espèces réactives de l'oxygène/métabolisme , Progéniteurs endothéliaux/métabolisme , Récepteurs à hydrocarbure aromatique/métabolisme , Cytochrome P-450 CYP1A1/métabolisme
7.
Int J Mol Sci ; 24(15)2023 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-37569446

RÉSUMÉ

This study investigated the protective effect of glutathione (GSH), an antioxidant drug, against doxorubicin (DOX)-induced cardiotoxicity. Human cardiac progenitor cells (hCPCs) treated with DOX (250 to 500 nM) showed increased viability and reduced ROS generation and apoptosis with GSH treatment (0.1 to 1 mM) for 24 h. In contrast to the 500 nM DOX group, pERK levels were restored in the group co-treated with GSH and suppression of ERK signaling improved hCPCs' survival. Similarly to the previous results, the reduced potency of hCPCs in the 100 nM DOX group, which did not affect cell viability, was ameliorated by co-treatment with GSH (0.1 to 1 mM). Furthermore, GSH was protected against DOX-induced cardiotoxicity in the in vivo model (DOX 20 mg/kg, GSH 100 mg/kg). These results suggest that GSH is a potential therapeutic strategy for DOX-induced cardiotoxicity, which performs its function via ROS reduction and pERK signal regulation.

9.
Exp Mol Med ; 53(9): 1423-1436, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34584195

RÉSUMÉ

Stem cell-based therapies with clinical applications require millions of cells. Therefore, repeated subculture is essential for cellular expansion, which is often complicated by replicative senescence. Cellular senescence contributes to reduced stem cell regenerative potential as it inhibits stem cell proliferation and differentiation as well as the activation of the senescence-associated secretory phenotype (SASP). In this study, we employed MHY-1685, a novel mammalian target of rapamycin (mTOR) inhibitor, and examined its long-term priming effect on the activities of senile human cardiac stem cells (hCSCs) and the functional benefits of primed hCSCs after transplantation. In vitro experiments showed that the MHY-1685‒primed hCSCs exhibited higher viability in response to oxidative stress and an enhanced proliferation potential compared to that of the unprimed senile hCSCs. Interestingly, priming MHY-1685 enhanced the expression of stemness-related markers in senile hCSCs and provided the differentiation potential of hCSCs into vascular lineages. In vivo experiment with echocardiography showed that transplantation of MHY-1685‒primed hCSCs improved cardiac function than that of the unprimed senile hCSCs at 4 weeks post-MI. In addition, hearts transplanted with MHY-1685-primed hCSCs exhibited significantly lower cardiac fibrosis and higher capillary density than that of the unprimed senile hCSCs. In confocal fluorescence imaging, MHY-1685‒primed hCSCs survived for longer durations than that of the unprimed senile hCSCs and had a higher potential to differentiate into endothelial cells (ECs) within the infarcted hearts. These findings suggest that MHY-1685 can rejuvenate senile hCSCs by modulating autophagy and that as a senescence inhibitor, MHY-1685 can provide opportunities to improve hCSC-based myocardial regeneration.


Sujet(s)
Autophagie , Différenciation cellulaire , Myoblastes cardiaques/cytologie , Myoblastes cardiaques/métabolisme , Régénération , Cellules souches/cytologie , Cellules souches/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Fibrose , Humains , Mâle , Myocarde/métabolisme , Myocarde/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Transduction du signal , Transplantation de cellules souches , Sérine-thréonine kinases TOR/métabolisme
10.
Exp Mol Med ; 52(4): 615-628, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32273566

RÉSUMÉ

The mammalian target of rapamycin (mTOR) signaling pathway efficiently regulates the energy state of cells and maintains tissue homeostasis. Dysregulation of the mTOR pathway has been implicated in several human diseases. Rapamycin is a specific inhibitor of mTOR and pharmacological inhibition of mTOR with rapamycin promote cardiac cell generation from the differentiation of mouse and human embryonic stem cells. These studies strongly implicate a role of sustained mTOR activity in the differentiating functions of embryonic stem cells; however, they do not directly address the required effect for sustained mTOR activity in human cardiac progenitor cells. In the present study, we evaluated the effect of mTOR inhibition by rapamycin on the cellular function of human cardiac progenitor cells and discovered that treatment with rapamycin markedly attenuated replicative cell senescence in human cardiac progenitor cells (hCPCs) and promoted their cellular functions. Furthermore, rapamycin not only inhibited mTOR signaling but also influenced signaling pathways, including STAT3 and PIM1, in hCPCs. Therefore, these data reveal a crucial function for rapamycin in senescent hCPCs and provide clinical strategies based on chronic mTOR activity.


Sujet(s)
Vieillissement de la cellule/effets des médicaments et des substances chimiques , Myoblastes cardiaques/effets des médicaments et des substances chimiques , Myoblastes cardiaques/métabolisme , Protéines proto-oncogènes c-pim-1/métabolisme , Facteur de transcription STAT-3/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Biologie informatique/méthodes , Analyse de profil d'expression de gènes , Humains , Sirolimus/pharmacologie , Cellules souches/métabolisme
11.
Tissue Eng Regen Med ; 17(3): 323-333, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32227286

RÉSUMÉ

BACKGROUND: Despite promising advances in stem cell-based therapy, the treatment of ischemic cardiovascular diseases remains a big challenge due to both the insufficient in vivo viability of transplanted cells and poor angiogenic potential of stem cells. The goal of this study was to develop therapeutic human cardiac progenitor cells (hCPCs) for ischemic cardiovascular diseases with a novel M13 peptide carrier. METHOD: In this study, an engineered M13 peptide carrier was successfully generated using a QuikChange Kit. The cellular function of M13 peptide carrier-treated hCPCs was assessed using a tube formation assay and scratch wound healing assay. The in vivo engraftment and cell survival bioactivities of transplanted cells were demonstrated by immunohistochemistry after hCPC transplantation into a myocardial infarction animal model. RESULTS: The engineered M13RGD+SDKP peptide carrier, which expressed RGD peptide on PIII site and SDKP peptide on PVIII site, did not affect morphologic change and proliferation ability in hCPCs. In contrast, hCPCs treated with M13RGD+SDKP showed enhanced angiogenic capacity, including tube formation and migration capacity. Moreover, transplanted hCPCs with M13RGD+SDKP were engrafted into the ischemic region and promoted in vivo cell survival. CONCLUSION: Our present data provides a promising protocol for CPC-based cell therapy via short-term cell priming of hCPCs with engineered M13RGD+SDKP before cell transplantation for treatment of cardiovascular disease.


Sujet(s)
Agents angiogéniques/pharmacologie , Infarctus du myocarde/thérapie , Peptides/métabolisme , Transplantation de cellules souches , Cellules souches/effets des médicaments et des substances chimiques , Animaux , Bactériophage M13/génétique , Maladies cardiovasculaires , Survie cellulaire , Cellules endothéliales , Génie génétique , Humains , Mâle , Souris de lignée BALB C , Myocytes cardiaques/transplantation , Peptides/pharmacologie , Cicatrisation de plaie
12.
Mar Drugs ; 17(6)2019 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-31234277

RÉSUMÉ

Cardiac progenitor cells (CPCs) are resident stem cells present in a small portion of ischemic hearts and function in repairing the damaged heart tissue. Intense oxidative stress impairs cell metabolism thereby decreasing cell viability. Protecting CPCs from undergoing cellular apoptosis during oxidative stress is crucial in optimizing CPC-based therapy. Histochrome (sodium salt of echinochrome A-a common sea urchin pigment) is an antioxidant drug that has been clinically used as a pharmacologic agent for ischemia/reperfusion injury in Russia. However, the mechanistic effect of histochrome on CPCs has never been reported. We investigated the protective effect of histochrome pretreatment on human CPCs (hCPCs) against hydrogen peroxide (H2O2)-induced oxidative stress. Annexin V/7-aminoactinomycin D (7-AAD) assay revealed that histochrome-treated CPCs showed significant protective effects against H2O2-induced cell death. The anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2) and Bcl-xL were significantly upregulated, whereas the pro-apoptotic proteins BCL2-associated X (Bax), H2O2-induced cleaved caspase-3, and the DNA damage marker, phosphorylated histone (γH2A.X) foci, were significantly downregulated upon histochrome treatment of hCPCs in vitro. Further, prolonged incubation with histochrome alleviated the replicative cellular senescence of hCPCs. In conclusion, we report the protective effect of histochrome against oxidative stress and present the use of a potent and bio-safe cell priming agent as a potential therapeutic strategy in patient-derived hCPCs to treat heart disease.


Sujet(s)
Myocytes cardiaques/effets des médicaments et des substances chimiques , Naphtoquinones/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Cellules souches/effets des médicaments et des substances chimiques , Annexine A5/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéines régulatrices de l'apoptose/métabolisme , Caspase-3/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques , Humains , Peroxyde d'hydrogène/pharmacologie , Myocytes cardiaques/métabolisme , Lésion d'ischémie-reperfusion/induit chimiquement , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Russie , Protéine Bax/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE