Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
4.
Stem Cell Res Ther ; 10(1): 352, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31779687

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a common neurotrauma leading to brain dysfunction and death. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) hold promise in the treatment of TBI. However, their efficacy is modest due to low survival and differentiation under the harsh microenvironment of the injured brain. MG53, a member of TRIM family protein, plays a vital role in cell and tissue damage repair. The present study aims to test whether MG53 preserves hUC-MSCs against oxidative stress and enhances stem cell survival and efficacy in TBI treatment. METHODS: In this study, we performed a series of in vitro and in vivo experiments in hUC-MSCs and mice to define the function of MG53 enhancing survival, neurogenesis, and therapeutic efficacy of stem cells in murine traumatic brain injury. RESULTS: We found that recombinant human MG53 (rhMG53) protein protected hUC-MSCs against H2O2-induced oxidative damage and stimulated hUC-MSC proliferation and migration. In a mouse model of contusion-induced TBI, intravenous administration of MG53 protein preserved the survival of transplanted hUC-MSCs, mitigated brain edema, reduced neurological deficits, and relieved anxiety and depressive-like behaviors. Co-treatment of MG53 and hUC-MSCs enhanced neurogenesis by reducing apoptosis and improving PI3K/Akt-GSK3ß signaling. CONCLUSION: MG53 enhances the efficacy of hUC-MSCs in the recovery of TBI, indicating that such adjunctive therapy may provide a novel strategy to lessen damage and optimize recovery for brain injury.


Subject(s)
Brain Injuries, Traumatic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Oxidative Stress , Signal Transduction , Tripartite Motif Proteins/metabolism , Umbilical Cord , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/therapy , Cell Survival , Disease Models, Animal , Heterografts , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Umbilical Cord/metabolism , Umbilical Cord/pathology
5.
Toxicol Lett ; 261: 49-58, 2016 Nov 02.
Article in English | MEDLINE | ID: mdl-27586268

ABSTRACT

Recently, we reported that ß-carotene exhibited anticancer activity against human esophageal squamous cell carcinoma cells in vitro. In the present study, we examined a novel therapeutic strategy by combining ß-carotene with 5-fluorouracil (5-FU) in human esophageal cancer in vitro and in vivo, and elucidated the underlying mechanisms. We found that the combination of 5-FU and ß-carotene displayed greater growth inhibitory effects than did either compound alone in esophageal squamous cell carcinoma (ESCC) cells. In addition, the combination of 5-FU and ß-carotene displayed greater tumor growth inhibition in an Eca109 xenograft mouse model than did a single agent with low systemic toxicity. ß-Carotene enhanced 5-FU-induced apoptosis. TUNEL staining revealed that the rate of TUNEL-positive cells was markedly increased in tumor tissues after treatment with 5-FU and ß-carotene. Western blotting and immunohistochemistry revealed the down-regulation of Bcl-2 and PCNA and the up-regulation of Bax and caspase-3 in tumor tissues. Further studies demonstrated that the combined administration of 5-FU and ß-carotene significantly down-regulated the protein levels of Cav-1, p-AKT, p-NF-κB, p-mTOR and p-p70S6K in Eca109 cells more effectively than did 5-FU alone. These data suggested that the combined therapy of 5-FU and ß-carotene exerted synergistic antitumor effects in vivo and in vitro and could constitute a novel therapeutic treatment for ESCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Fluorouracil/therapeutic use , beta Carotene/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Therapy, Combination , Fluorouracil/administration & dosage , Humans , Mice , Neoplasms, Experimental/drug therapy , beta Carotene/administration & dosage
6.
Mol Cells ; 39(5): 418-25, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27109421

ABSTRACT

Resveratrol (RES) plays a critical role in the fate of cells and longevity of animals via activation of the sirtuins1 (SIRT1) gene. In the present study, we intend to investigate whether RES could promote the self-renewal and neural-lineage differentiation in human umbilical cord derived MSCs (hUC-MSCs) in vitro at concentrations ranging from 0.1 to 10 µM, and whether it exerts the effects by modulating the SIRT1 signaling. Herein, we demonstrated that RES at the concentrations of 0.1, 1 and 2.5 µM could promote cell viability and proliferation, mitigate senescence and induce expression of SIRT1 and Proliferating Cell Nuclear Antigen (PCNA) while inhibit the expression of p53 and p16. However, the effects were reversed by 5 and 10 µM of RES. Furthermore, RES could promote neural differentiation in a dose-dependent manner as evidenced by morphological changes and expression of neural markers (Nestin, ßIII-tubulin and NSE), as well as pro-neural transcription factors Neurogenin (Ngn)1, Ngn2 and Mash1. Taken together, RES exerts a dosage-dependent effect on the self-renewal and neural differentiation of hUC-MSCs via SIRT1 signaling. The current study provides a new strategy to regulate the fate of hUC-MSCs and suggests a more favorable in vitro cell culture conditions for hUC-MSCs-based therapies for some intractable neurological disorders.


Subject(s)
Mesenchymal Stem Cells/drug effects , Neurons/drug effects , Sirtuin 1/metabolism , Stilbenes/pharmacology , Umbilical Cord/cytology , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Mesenchymal Stem Cells/cytology , Neurons/cytology , Proliferating Cell Nuclear Antigen/metabolism , Resveratrol , Signal Transduction/drug effects
7.
J Biochem Mol Toxicol ; 30(3): 148-57, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26733226

ABSTRACT

ß-carotene, a type of terpenoid, has many metabolic and physiological functions. In particular, ß-carotene has an antitumor effect. However, the efficacy of ß-carotene against esophageal squamous cell carcinoma (ESCC) remains unclear. In our study, ß-carotene inhibited the growth of ESCC cells and downregulated expression of the Caveolin-1 (Cav-1) protein. Cav-1 protein was expressed only in ESCC cells, not in Het-1A cells. Moreover, ß-carotene triggered apoptosis, induced cell cycle G0/G1 phase arrest, and inhibited cell migration. To explore the mechanism involved in these processes, we further examined the effect of ß-carotene on the Cav-1-mediated AKT/NF-κB pathway. The results showed that the level of AKT and NF-κB phosphorylation was dramatically inhibited, which led to an increase in the Bax/Bcl-2 ratio. Correspondingly, the activity of Caspase-3 was also enhanced. These data suggest that ß-carotene has an antiproliferative role in ESCC cells and may be a promising chemotherapeutic agent for use against ESCC cells.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Caveolin 1/metabolism , Esophageal Neoplasms/pathology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , beta Carotene/pharmacology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...