Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cytotherapy ; 24(4): 421-427, 2022 04.
Article in English | MEDLINE | ID: mdl-35086778

ABSTRACT

BACKGROUND AIMS: To explore the long-term safety and benefit of umbilical cord mesenchymal stromal cell (MSCs) plus autologous bone marrow mononuclear cell (aBM-MNC) stem cell transplantation (SCT) without immunotherapy in established type 1 diabetes (T1D). METHODS: In the primary completion of this trial (ClinicalTrials.gov identifier: NCT01374854), the authors randomized patients (n = 21 per group) to either SCT or standard care (control) and previously reported effects on insulin secretion. The authors report about the incidence of chronic diabetes complications (primary endpoint) after 8 years of follow-up. The authors also report on secondary endpoints, safety, islet function and metabolic control. RESULTS: Data were obtained from 14 of 21 patients in the SCT group and 15 of 21 patients in the control group who completed follow-up. At 8 years, the incidence of peripheral neuropathy was 7.1% (one of 14) in the SCT group versus 46.7% (seven of 15) in the control group (P = 0.017). The incidence of diabetic nephropathy was 7.1% (one of 14) in the SCT group versus 40.0% (six of 15) in the control group (P = 0.039). The incidence of retinopathy was 7.1% (one of 14) in the SCT group versus 33.3% (five of 15) in the control group (P = 0.081). Two patients (two of 14, 14.3%) in the SCT group and 11 patients (11 of 15, 73.3%) in the control group developed at least one complication (P = 0.001). One and six patients in the SCT group and control group, respectively, had at least two complications (P = 0.039). No malignancies were reported in the treated group. CONCLUSIONS: Co-transplantation of umbilical cord MSCs and aBM-MNCs in patients with established T1D was associated with reduced incidence of chronic diabetes complications.


Subject(s)
Diabetes Complications , Diabetes Mellitus, Type 1 , Graft vs Host Disease , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Bone Marrow , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/therapy , Follow-Up Studies , Humans , Mesenchymal Stem Cell Transplantation/adverse effects , Pilot Projects , Umbilical Cord
2.
Stem Cell Res Ther ; 11(1): 97, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32127037

ABSTRACT

BACKGROUND: Hypoxia is a major cause of beta cell death and dysfunction after transplantation. The aim of this study was to investigate the effect of exosomes derived from mesenchymal stem cells (MSCs) on beta cells under hypoxic conditions and the potential underlying mechanisms. METHODS: Exosomes were isolated from the conditioned medium of human umbilical cord MSCs and identified by WB, NTA, and transmission electron microscopy. Beta cells (ßTC-6) were cultured in serum-free medium in the presence or absence of exosomes under 2% oxygen conditions. Cell viability and apoptosis were analysed with a CCK-8 assay and a flow cytometry-based annexin V-FITC/PI apoptosis detection kit, respectively. Endoplasmic reticulum stress (ER stress) proteins and apoptosis-related proteins were detected by the WB method. MiRNAs contained in MSC exosomes were determined by Illumina HiSeq, and treatment with specific miRNA mimics or inhibitors of the most abundant miRNAs was used to reveal the underlying mechanism of exosomes. RESULTS: Exosomes derived from MSC-conditioned culture medium were 40-100 nm in diameter and expressed the exosome markers CD9, CD63, CD81, HSP70, and Flotillin 1, as well as the MSC markers CD73, CD90, and CD105. Hypoxia significantly induced beta cell apoptosis, while MSC exosomes remarkably improved beta cell survival. The WB results showed that ER stress-related proteins, including GRP78, GRP94, p-eIF2α and CHOP, and the apoptosis-related proteins cleaved caspase 3 and PARP, were upregulated under hypoxic conditions but were inhibited by MSC exosomes. Moreover, the p38 MAPK signalling pathway was activated by hypoxia and was inhibited by MSC exosomes. The Illumina HiSeq results show that MSC exosomes were rich in miR-21, let-7 g, miR-1246, miR-381, and miR-100. After transfection with miRNA mimics, the viability of beta cells under hypoxia was increased significantly by miR-21 mimic, and the p38 MAPK and ER stress-related proteins in beta cells were downregulated. These changes were reversed after exosomes were pretreated with miR-21 inhibitor. CONCLUSIONS: Exosomes derived from MSCs could protect beta cells against apoptosis induced by hypoxia, largely by carrying miR-21, alleviating ER stress and inhibiting p38 MAPK signalling. This result indicated that MSC exosomes might improve encapsulated islet survival and benefit diabetes patients.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Apoptosis , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Exosomes/metabolism , Humans , Hypoxia/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphorylation , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
J Exp Clin Cancer Res ; 38(1): 150, 2019 Apr 08.
Article in English | MEDLINE | ID: mdl-30961642

ABSTRACT

BACKGROUND: Deregulated ErbB signaling plays an important role in tumorigenesis of pancreatic cancer. However, patients with pancreatic cancer benefit little from current existed therapies targeting the ErbB signaling. Here, we explore the potential anti-tumor activity of Valproic acid against pancreatic cancer via targeting ErbB family members. METHODS: Cell viability assay and apoptosis evaluation were carried out to determine the efficacy of VPA on pancreatic cancer cells. Western blot analyses were performed to determine the expression and activation of proteins. Apoptosis enzyme-linked immunosorbent assay was used to quantify cytoplasmic histone associated DNA fragments. Lentiviral expression system was used to introduce overexpression of exogeneous genes or gene-targeting short hairpin RNAs (shRNAs). qRT-PCR was carried out to analyze the mRNAs and miRNAs expression levels. Tumor xenograft model was established to evaluate the in vivo anti-pancreatic cancer activity of VPA. RESULTS: VPA preferentially inhibited cell proliferation/survival of, and induced apoptosis in EGFR/ErbB2/ErbB3-coexpressing pancreatic cancer cells within its clinically achievable range [40~100 mg/L (0.24~0.6 mmol/L)]. Mechanistic investigations revealed that VPA treatment resulted in simultaneous significant down-regulation of EGFR, ErbB2, and ErbB3 in pancreatic cancer cells likely via induction of ErbB family members-targeting microRNAs. Moreover, the anti-pancreatic cancer activity of VPA was further validated in tumor xenograft model. CONCLUSIONS: Our data strongly suggest that VPA may be added to the treatment regimens for pancreatic cancer patients with co-overexpression of the ErbB family members.


Subject(s)
MicroRNAs/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Receptor, ErbB-3/metabolism , Valproic Acid/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Signal Transduction , Valproic Acid/pharmacology
4.
Oxid Med Cell Longev ; 2019: 2746521, 2019.
Article in English | MEDLINE | ID: mdl-30809321

ABSTRACT

The aged kidney is susceptible to acute injury due presumably to its decreased ability to handle additional challenges, such as endoplasmic reticulum (ER) stress. This was tested by giving tunicamycin, an ER stress inducer, to either old or young mice. Injection of high dose caused renal failure in old mice, not in young mice. Moreover, injection of low dose resulted in severe renal damage in old mice, confirming the increased susceptibility of aged kidney to ER stress. There existed an abnormality in ER stress response kinetics in aged kidney, characterized by a loss of XBP-1 splicing and decreased PERK-eIF2α phosphorylation at late time point. The presence of excessive oxidative stress in aged kidney may play a role since high levels of oxidation increased ER stress-induced cell death and decreased IRE1 levels and XBP-1 splicing. Importantly, treatment with antioxidants protected old mice from kidney injury and normalized IRE1 and XBP-1 responses. Furthermore, older mice (6 months old) transgenic with antioxidative stress AGER1 were protected from ER stress-induced kidney injury. In conclusion, the decreased ability to handle ER stress, partly due to the presence of excessive oxidative stress, may contribute to increased susceptibility of the aging kidney to acute injury.


Subject(s)
Acute Kidney Injury/pathology , Aging/pathology , Endoplasmic Reticulum Stress , Oxidative Stress , Alternative Splicing/drug effects , Animals , Antioxidants/metabolism , Endoplasmic Reticulum Stress/drug effects , Female , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tunicamycin/toxicity , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , X-Box Binding Protein 1/metabolism
5.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 26(6): 1858-1862, 2018 Dec.
Article in Chinese | MEDLINE | ID: mdl-30501734

ABSTRACT

Mesenchymal stem cells (MSC) have the potential of multi-directional differentiation, and can recruit endothelial cells, promote their proliferation, migration and angiogenesis, improve blood perfusion and oxygen suppliment, and repair damaged tissue. Exosome secreted by MSC contain mother cell-specific proteins, lipids and nucleic acids, and acts as signaling molecule, playing an important role in cell communication, thereby altering target cell function. In this review, the biological characteristics of MSC and its exosome, the mechanism of promoting vascular regeneration in patients with ischemic diseases, and the mechanism of hypoxia-inducible factor-1α(HIF-1α) in the vascular ischemia of ischemic diseases are all summarized briefly.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Cell Differentiation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Neovascularization, Pathologic , Neovascularization, Physiologic
6.
Biochem Biophys Res Commun ; 501(1): 212-219, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29715459

ABSTRACT

We had previously demonstrated that increased expression of ErbB3 is required for ErbB2-mediated paclitaxel resistance in breast cancer cells. In the present study, we have explored the possible role of mesenchymal stem cells (MSCs) in regulating the paclitaxel-sensitivity of ErbB2/ErbB3-coexpressing breast cancer cells. We show that human umbilical cord-derived MSCs express significantly higher level of neuregulin-1 as compared with ErbB2/ErbB3-coexpressing breast cancer cells themselves. Coculture or treatment with conditioned medium of MSCs not only decreases the anti-proliferation effect of paclitaxel on ErbB2/ErbB3-coexpressing breast cancer cells, but also significantly inhibits paclitaxel-induced apoptosis. We further demonstrate that this MSCs-drived paclitaxel resistance in ErbB2/ErbB3-coexpressing breast cancer cells could be attributed to upregulation of Survivin via paracrine effect of NRG-1/ErbB3/PI-3K/Akt signaling, as either specific knockdown expression of ErbB3, or blocking of downstream PI-3K/Akt signaling, or specific inhibition of Survivin can completely reverse this effect. Moreover, targeted knockdown of NRG-1 expression in MSCs abrogates theirs effect on paclitaxel sensitivity of ErbB2/ErbB3-coexpressing breast cancer cells. Taken together, our study indicate that paracrine of NRG-1 by MSCs induces paclitaxel resistance in ErbB2/ErbB3-coexpressing breast cancer cells through PI-3K/Akt signaling-dependent upregulation of Survivin. Our findings suggest that simultaneously targeting mesenchymal stem cells in tumor microenvironment may be a novel strategy to overcome paclitaxel resistance in patients with ErbB2/ErbB3-coexpressing breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Neuregulin-1/metabolism , Paclitaxel/pharmacology , Breast Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Gene Knockdown Techniques , Genes, erbB-2 , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Neuregulin-1/antagonists & inhibitors , Neuregulin-1/genetics , Paracrine Communication , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Survivin
7.
PLoS One ; 12(8): e0183680, 2017.
Article in English | MEDLINE | ID: mdl-28841673

ABSTRACT

C/EBP-homologous protein (CHOP) is an important component of the endoplasmic reticulum (ER) stress response. We demonstrated the induction of ER stress in response to tunicamycin stimulation, as evidenced by increased expression of chaperone proteins Grp78, Grp94, and enhanced eukaryotic initiation factor 2 subunit 1 (eIF2α) phosphorylation in hepatocellular carcinoma cells. Tunicamycin-induced ER stress resulted in apoptosis and autophagy simultaneously. While inhibition of autophagy mediated by 3-methyladenine pretreatment or direct knockdown of LC3B promoted cell apoptosis, activation of autophagy with rapamycin decreased tunicamycin- induced apoptosis in HCC cells. Furthermore, CHOP was shown to be significantly upregulated upon treatment with tunicamycin in HCC cells. Specific knockdown of CHOP not only enhanced tunicamycin-induced autophagy, but also significantly attenuated ER stress-induced apoptosis in HCC cells. Accordingly, simultaneous inhibition of autophagy in HCC cells with CHOP-knockdown could partially resensitize ER stress-induced apoptosis. Taken together, our data indicate that CHOP may favor ER stress-induced apoptosis in HCC cells via inhibition of autophagy in vitro.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Carcinoma, Hepatocellular/pathology , Endoplasmic Reticulum Stress/physiology , Liver Neoplasms/pathology , Transcription Factor CHOP/physiology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Liver Neoplasms/metabolism , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
Mol Med Rep ; 15(6): 4041-4048, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28487936

ABSTRACT

Ectopic expression of musculo aponeurotic fibrosarcoma BZIP transcription factor (Maf) A, has previously been demonstrated to induce insulin expression in non­ß­cell lines. Protein transduction domains acting as an alternative delivery strategy may deliver heterogeneous proteins into cells. A sequence of 11 arginine residues (11R) has been demonstrated to act as a particularly efficient vector to introduce proteins into various cell types. The present study constructed 11R­fused MafA to achieve transduction of the protein into cellular membranes and subsequently examined the therapeutic effect of the MafA­11R protein in streptozotocin­induced diabetes. A small animal imaging system was used to demonstrate that 11R introduced proteins into cells. The MafA­11R protein was then injected into the tale vein of healthy male mice, and western blot analysis and immunofluorescence staining was performed to identify the location of the recombinant protein. Ameliorated hyperglycemia in the MafA­11R­treated diabetic mice was demonstrated via the improved intraperitoneal glucose tolerance test (IPGTT) and glucose­stimulated insulin release. Furthermore, insulin producing cells were detected in the jejunum of the MafA­11R treated mice. The results of the present study indicated that MafA­11R delivery may act as a novel and potential therapeutic strategy for the future and will not present adverse effects associated with viral vector­mediated gene therapies.


Subject(s)
Basic-Leucine Zipper Transcription Factors/pharmacology , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/blood , Peptides , Recombinant Fusion Proteins/pharmacology , Animals , Basic-Leucine Zipper Transcription Factors/chemistry , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line , Cell Survival/drug effects , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Glucose Tolerance Test , Insulin/blood , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Mice , Peptides/chemistry , Recombinant Fusion Proteins/genetics , Tissue Distribution
9.
Stem Cells Dev ; 26(17): 1283-1292, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28548571

ABSTRACT

RHO GTPases regulate cell migration, cell-cycle progression, and cell survival in response to extracellular stimuli. However, the regulatory effects of RHO GTPases in mesenchymal stromal cells (MSCs) are unclear. Herein, we show that CDC42 acts as an essential factor in regulating cell proliferation and also takes part in lipotoxic effects of palmitate in human umbilical cord Wharton's jelly derived MSCs (hWJ-MSCs). Cultured human bone marrow, adipose tissue, and hWJ-MSC derived cells had varying pro-inflammatory cytokine secretion levels and cell death rates when treated by palmitate. Strikingly, the proliferation rate of these types of MSCs correlated with their sensitivity to palmitate. A glutathione-S-transferase pull-down assay demonstrated that hWJ-MSCs had the highest activation of CDC42, which was increased by palmitate treatment in a time-dependent manner. We demonstrated that palmitate-induced synthesis of pro-inflammatory cytokines and cell death was attenuated by shRNA against CDC42. In CDC42 depleted hWJ-MSCs, population-doubling levels were notably decreased, and phosphorylation of ERK1/2 and p38 MAPK was reduced. Our data therefore suggest a mechanistic role for CDC42 activity in hWJ-MSC proliferation and identified CDC42 activity as a promising pharmacological target for ameliorating lipotoxic cell dysfunction and death.


Subject(s)
Mesenchymal Stem Cells/cytology , Palmitates/toxicity , Umbilical Cord/cytology , Wharton Jelly/cytology , cdc42 GTP-Binding Protein/metabolism , Adult , Cell Death/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , MAP Kinase Signaling System/drug effects , Male , Mesenchymal Stem Cells/drug effects , Middle Aged
10.
Exp Ther Med ; 11(6): 2425-2431, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27284330

ABSTRACT

The aim of the present study was to investigate the ability of bone marrow-derived mesenchymal stem cells (BMSCs) to repair radiation-induced acute intestinal injury, and to elucidate the underlying repair mechanism. Male Sprague-Dawley rats were subjected to whole abdominal irradiation using a single medical linear accelerator (12 Gy) and randomly assigned to two groups. Rats in the BMSC-treated group were injected with 1 ml BMSC suspension (2×106 cells/ml) via the tail vein, while the control group rats were injected with normal saline. BMSCs were identified by detecting the expression of CD29, CD90, CD34 and CD45 using flow cytometry. The expression of the cytokines stromal cell-derived factor 1 (SDF-1), prostaglandin E2 (PGE2) and interleukin (IL)-2 was detected using immunohistochemical techniques. Plasma citrulline concentrations were evaluated using an ELISA kit. Rat general conditions, including body weight, and changes in cellular morphology were also recorded. The results suggested that BMSCs exerted a protective effect on radiation-induced acute intestinal injury in rats. The histological damage was rapidly repaired in the BMSC-treated group. In addition, the BMSC-treated group showed significantly reduced radiation injury scores (P<0.01), mildly reduced body weight and plasma citrulline levels, significantly more rapid recovery (P<0.01), significantly reduced expression of the cytokines PGE2 and IL-2 (P<0.05) and significantly increased SDF-1 expression (P<0.01) compared with the control group. In summary, the present results indicate that BMSCs are able to effectively reduce inflammation and promote repair of the structure and function of intestinal tissues damaged by radiation exposure, suggesting that they may provide a promising therapeutic agent.

11.
Oncotarget ; 7(25): 37693-37713, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27177222

ABSTRACT

Elevated expression of Survivin correlates with poor prognosis, tumor recurrence, and drug resistance in various human cancers, including non-small cell lung cancer (NSCLC). The underlying mechanism of Survivin upregulation in cancer cells remains elusive. To date, no Survivin-targeted therapy has been approved for cancer treatment. Here, we explored the molecular basis resulting in Survivin overexpression in NSCLC and investigated the antitumor activity of the class I HDAC inhibitor entinostat in combination with paclitaxel. Our data showed that entinostat significantly enhanced paclitaxel-mediated anti-proliferative/anti-survival effects on NSCLC cells in vitro and in vivo. Mechanistically, entinostat selectively decreased expression of Survivin via induction of miR-203 (in vitro and in vivo) and miR-542-3p (in vitro). Moreover, analysis of NSCLC patient samples revealed that the expression levels of miR-203 were downregulated due to promoter hypermethylation in 45% of NSCLC tumors. In contrast, increased expression of both DNA methytransferase I (DNMT1) and Survivin was observed and significantly correlated with the reduced miR-203 in NSCLC. Collectively, these data shed new lights on the molecular mechanism of Survivin upregulation in NSCLC. Our findings also support that the combinatorial treatments of entinostat and paclitaxel will likely exhibit survival benefit in the NSCLC patients with overexpression of DNMT1 and/or Survivin. The DNMT1-miR-203-Survivin signaling axis may provide a new avenue for the development of novel epigenetic approaches to enhance the chemotherapeutic efficacy against NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Paclitaxel/pharmacology , Apoptosis , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Drug Synergism , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Pyridines/pharmacology , Survivin
12.
Diabetes Care ; 39(1): 149-57, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26628416

ABSTRACT

OBJECTIVE: To determine the safety and effects on insulin secretion of umbilical cord (UC) mesenchymal stromal cells (MSCs) plus autologous bone marrow mononuclear cell (aBM-MNC) stem cell transplantation (SCT) without immunotherapy in established type 1 diabetes (T1D). RESEARCH DESIGN AND METHODS: Between January 2009 and December 2010, 42 patients with T1D were randomized (n = 21/group) to either SCT (1.1 × 10(6)/kg UC-MSC, 106.8 × 10(6)/kg aBM-MNC through supraselective pancreatic artery cannulation) or standard care (control). Patients were followed for 1 year at 3-month intervals. The primary end point was C-peptide area under the curve (AUC(C-Pep)) during an oral glucose tolerance test at 1 year. Additional end points were safety and tolerability of the procedure, metabolic control, and quality of life. RESULTS: The treatment was well tolerated. At 1 year, metabolic measures improved in treated patients: AUCC-Pep increased 105.7% (6.6 ± 6.1 to 13.6 ± 8.1 pmol/mL/180 min, P = 0.00012) in 20 of 21 responders, whereas it decreased 7.7% in control subjects (8.4 ± 6.8 to 7.7 ± 4.5 pmol/mL/180 min, P = 0.013 vs. SCT); insulin area under the curve increased 49.3% (1,477.8 ± 1,012.8 to 2,205.5 ± 1,194.0 mmol/mL/180 min, P = 0.01), whereas it decreased 5.7% in control subjects (1,517.7 ± 630.2 to 1,431.7 ± 441.6 mmol/mL/180 min, P = 0.027 vs. SCT). HbA1c decreased 12.6% (8.6 ± 0.81% [70.0 ± 7.1 mmol/mol] to 7.5 ± 1.0% [58.0 ± 8.6 mmol/mol], P < 0.01) in the treated group, whereas it increased 1.2% in the control group (8.7 ± 0.9% [72.0 ± 7.5 mmol/mol] to 8.8 ± 0.9% [73 ± 7.5 mmol/mol], P < 0.01 vs. SCT). Fasting glycemia decreased 24.4% (200.0 ± 51.1 to 151.2 ± 22.1 mg/dL, P < 0.002) and 4.3% in control subjects (192.4 ± 35.3 to 184.2 ± 34.3 mg/dL, P < 0.042). Daily insulin requirements decreased 29.2% in only the treated group (0.9 ± 0.2 to 0.6 ± 0.2 IU/day/kg, P = 0.001), with no change found in control subjects (0.9 ± 0.2 to 0.9 ± 0.2 IU/day/kg, P < 0.01 vs. SCT). CONCLUSIONS: Transplantation of UC-MSC and aBM-MNC was safe and associated with moderate improvement of metabolic measures in patients with established T1D.


Subject(s)
Bone Marrow Transplantation , Diabetes Mellitus, Type 1/therapy , Mesenchymal Stem Cell Transplantation , Adolescent , Adult , C-Peptide/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/drug therapy , Female , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin/therapeutic use , Insulin Secretion , Male , Pilot Projects , Quality of Life , Transplantation, Autologous , Umbilical Cord/cytology , Young Adult
13.
Mol Med Rep ; 12(5): 7065-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26323987

ABSTRACT

The present study aimed to observe the role of mesenchymal stem cells (MSCs) in the repair of acute skin damage caused by radiation. Rat bone marrow MSCs (BMSCs) were isolated and cultured in vitro. A rat model of radiation­induced acute skin damage was established by irradiation of the hind legs of Sprague-Dawley rats using a linear accelerator (45 Gy). After irradiation, rats were randomly divided into two groups: BMSC group and control group. Rats in the BMSC group were treated with a tail vein injection of 2x106 BMSCs (1 ml) immediately after irradiation and a local multipoint injection of 2x106 BMSCs at the injured area two weeks later. Then the wound healing of each rat was observed. The expression of transforming growth factor (TGF)­ß1, stromal cell­derived factor-1 (SDF­1) and prostaglandin E2 (PGE2) in the wounded tissues was determined by immunohistochemistry. The results demonstrated that skin damage was milder in the BMSC group than in the control group. Moreover, the speed of healing in the BMSC group was better than that in the control group. In addition, the wound score, it was significantly lower in the BMSC group than in the control group (P<0.05). The expression of PGE2 and TGF­ß1 in the BMSC group was also significantly lower than that in the control group (P<0.05), whereas the SDF­1 expression was significantly higher in the BMSC group than that in the control group (P<0.05). BMSCs can effectively reduce inflammation and fibrosis in the wounded skin and promote the repair of acute radioactive skin injury. Thus, may be developed as a novel treatment for wound healing.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Skin/injuries , Wound Healing , Animals , Cell Differentiation , Cells, Cultured , Chemokine CXCL12/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Gamma Rays , Immunohistochemistry , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Skin/metabolism , Skin/pathology , Transforming Growth Factor beta1/metabolism
14.
Biochem Biophys Res Commun ; 453(3): 405-10, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25268319

ABSTRACT

The direct conversion of one cell type to another without an intermediate pluripotent stage is required for regenerative therapies. The ventral pancreas and liver share a common developmental origin. Recent studies have shown that hepatocytes could be induced to transdifferentiate into insulin-producing cells. In this paper, we showed a new strategy to achieve the direct conversion of human hepatocytes into surrogate ß cells. Hepatocytes were transfected with microRNA-302 (miR-302) mimic and Pdx1, Ngn3 and MafA expressed plasmids, followed by a chemical-defined culture system for maturation of insulin-secreting cells. Co-transfection of miR-302 mimic increased the transcription of pancreatic development-related genes (Sox17, Foxa2, and endogenous Pdx1). Furthermore, at the end of this treatment, hepatocytes became insulin expressed cells that released the hormone in response to a physiological glucose change in vitro. This work shows that miR-302 participation may facilitates the conversion of adult hepatocytes into pancreatic islets-like cells.


Subject(s)
Cell Differentiation/physiology , Hepatocytes/cytology , Islets of Langerhans/cytology , MicroRNAs/physiology , Base Sequence , Cells, Cultured , Culture Media , DNA Primers , Humans , MicroRNAs/genetics , Polymerase Chain Reaction
15.
Cytotherapy ; 16(4): 471-84, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24119646

ABSTRACT

BACKGROUND AIMS: Minimal change nephrotic syndrome is the most frequent cause of nephrotic syndrome in childhood. Current treatment regimes, which include glucocorticoid hormones and immunosuppressive therapy, are effective and have fast response. However, because of the side effects, long treatment course, poor patient compliance and relapse, novel approaches for the disease are highly desired. METHODS: The adriamycin-induced nephrotic rat model was established. Rats were allocated to a model group, a prednisone group or mesenchymal stromal cell (MSC) group. Clinical parameters in each treatment group were determined at 2 weeks, 4 weeks and 8 weeks. The messenger RNA (mRNA) levels of synaptopodin, p21 and monocyte chemoattractant protein-1 were determined through the use of quantitative real-time-polymerase chain reaction. Protein levels were determined by means of Western blot or enzyme-linked immunosorbent assay. Podocytes were isolated and apoptotic rate after adriamycin with or without MSC treatment was analyzed by means of flow cytometry. RESULTS: MSC intervention improved renal function as assessed by urinary protein, blood creatinine and triglyceride levels. MSC intervention reduced adriamycin-induced renal tissue damage visualized by immunohistochemistry and light and electron microscopic analysis and reduced adriamycin-induced podocyte apoptosis. After MSC intervention, mRNA and protein levels of synaptopodin and p21 in renal cortex were significantly increased. MSCs also restored synaptopodin mRNA and protein expression in isolated podocytes. In addition, monocyte chemoattractant protein-1 mRNA in renal cortex and protein level in serum of the MSC treatment group were significantly decreased compared with that in the adriamycin-induced nephropathy model group. CONCLUSIONS: Our data indicate that MSCs could protect rats from adriamycin-induced minimal change nephrotic syndrome, and the protective effects of MSCs are mediated through multiple actions.


Subject(s)
Kidney/drug effects , Mesenchymal Stem Cell Transplantation , Nephrosis, Lipoid/pathology , Nephrosis, Lipoid/therapy , Animals , Chemokine CCL2/biosynthesis , Doxorubicin/toxicity , Gene Expression Regulation , Humans , Kidney/pathology , Mesenchymal Stem Cells/cytology , Microfilament Proteins/biosynthesis , Nephrosis, Lipoid/chemically induced , Prednisone/administration & dosage , RNA, Messenger/biosynthesis , Rats , rho GTP-Binding Proteins/biosynthesis
16.
Cytotherapy ; 16(2): 258-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24290656

ABSTRACT

BACKGROUND AIMS: The use of bone marrow mononuclear cells (BM-MNCs) has achieved great outcomes in clinical practice. We aim to evaluate the efficacy and safety of autologous BM-MNC infusion and hyperbaric oxygen therapy (HOT) in type 2 diabetes mellitus. METHODS: This single-center, randomized, open-label, controlled clinical trial with a factorial design included two phases. The patients received standard medical therapy in the run-in phase; in the treatment phase, patients with glycated hemoglobin of 7.5-9.5% were randomly assigned into four groups and underwent BM-MNC infusion along with HOT (BM-MNC+HOT group), BM-MNC infusion (BM-MNC group), HOT (HOT group) and standard medical therapy (control group), respectively. The area under the curve of C-peptide was recorded as a primary end point. Our research is registered at ClinicalTrials.gov (NCT00767260). RESULTS: A total of 80 patients completed the follow-up. At 12 months after treatment, the area under the curve of C-peptide (ng/mL per 180 min) of the BM-MNC+HOT group and the BM-MNC group were significantly improved (34.0% and 43.8% from the baseline, respectively). The changes were both significant compared with that in the control group, but no remarkable change was observed in the HOT group. Treatment-related adverse events were mild, including transient abdominal pain (n = 5) and punctual hemorrhage (n = 3). CONCLUSIONS: BM-MNC infusion for type 2 diabetes mellitus improves islet function and metabolic control, with mild adverse effects. HOT does not synergize with BM-MNC infusion.


Subject(s)
Bone Marrow Cells/metabolism , Cell Transplantation , Diabetes Mellitus, Type 2/therapy , Hyperbaric Oxygenation , Insulin-Secreting Cells/metabolism , Leukocytes, Mononuclear/transplantation , Aged , Bone Marrow Cells/pathology , Cells, Cultured , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Insulin-Secreting Cells/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Treatment Outcome
17.
Sheng Li Xue Bao ; 65(6): 600-6, 2013 Dec 25.
Article in Chinese | MEDLINE | ID: mdl-24343717

ABSTRACT

The saturated free fatty acid (FFA), palmitate, could induce apoptosis in various cell types, but little is known about its effects on human umbilical cord-derived mesenchymal stem cells (hUC-MSCs). Here, we investigated whether palmitate induced apoptosis and endoplasmic reticulum (ER) stress in hUC-MSCs. hUC-MSCs were stained by labeled antibodies and identified by flow cytometry. After administration with palmitate, apoptotic cell was assessed by flow cytometry using the Annexin V-FITC/7-AAD apoptosis detection kit. Relative spliced XBP1 levels were analyzed using semi-quantitative RT-PCR. The mRNA of BiP, GRP94, ATF4 and CHOP were analyzed by real-time PCR. Relative BiP and CHOP protein were analyzed using Western blot analysis. The results showed that hUC-MSCs were homogeneously positive for MSC markers; palmitate increased apoptosis of hUC-MSCs and activated XBP1 splicing, BiP, GRP94, ATF4 and CHOP transcription. These findings suggest that palmitate induces apoptosis and ER stress in hUC-MSCs.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Mesenchymal Stem Cells/drug effects , Palmitates/pharmacology , Activating Transcription Factor 4/metabolism , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/cytology , Regulatory Factor X Transcription Factors , Transcription Factor CHOP/metabolism , Transcription Factors/metabolism , Umbilical Cord/cytology , X-Box Binding Protein 1
18.
BMC Cell Biol ; 14: 18, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23510080

ABSTRACT

BACKGROUND: The objective of this study was to evaluate the effect of bone marrow mesenchymal stem cells (BMSCs) on the apoptosis of granulosa cells (GCs) in rats. RESULTS: Cisplatin increased GC apoptosis from 0.59% to 13.04% in the control and cisplatin treatment groups, respectively, which was significantly reduced upon co-culture with BMSCs to 4.84%. Cisplatin treatment increased p21 and bax and decreased c-myc mRNA expression, which was reversed upon co-culture with BMSCs. As compared to young rats, increased apoptosis was observed in the perimenopausal rats (P < 0.001). After 3 months, the apoptosis rate in the BMSC group was significantly lower than that of the control group (P = 0.007). CONCLUSIONS: BMSC therapy may protect against GC apoptosis induced by cisplatin and perimenopause. Further studies are necessary to evaluate therapeutic efficacy of BMSCs.


Subject(s)
Bone Marrow Cells/drug effects , Cisplatin/administration & dosage , Coculture Techniques/methods , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Granulosa Cells/drug effects , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Perimenopause/physiology , Animals , Apoptosis/drug effects , Bone Marrow Cells/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Estrogens/administration & dosage , Female , Gene Expression Regulation/drug effects , Granulosa Cells/physiology , Mesenchymal Stem Cells/physiology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
19.
Endocrinology ; 153(11): 5275-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22968644

ABSTRACT

Elevated circulating saturated fatty acids concentration is commonly associated with poorly controlled diabetes. The highly prevalent free fatty acid palmitate could induce apoptosis in various cell types, but little is known about its effects on human mesenchymal stem cells (MSCs). Here, we report that prolonged exposure to palmitate induces human bone marrow-derived MSC (hBM-MSC) and human umbilical cord-derived MSC apoptosis. We investigated the role of endoplasmic reticulum (ER) stress, which is known to promote cell apoptosis. Palmitate activated XBP1 splicing, elF2α (eukaryotic translation initiation factor 2α) phosphorylation, and CHOP, ATF4, BiP, and GRP94 transcription in hBM-MSCs. ERK1/2 and p38 MAPK phosphorylation were also induced by palmitate in hBM-MSCs. A selective p38 inhibitor inhibited palmitate activation of the ER stress, whereas the ERK1/2 inhibitors had no effect. The AMP-activated protein kinase activator aminoimidazole carboxamide ribonucleotide blocked palmitate-induced ER stress and apoptosis. These findings suggest that palmitate induces ER stress and ERK1/2 and p38 activation in hBM-MSCs, and AMP-activated protein kinase activator prevents the deleterious effects of palmitate by inhibiting ER stress and apoptosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Mesenchymal Stem Cells/drug effects , Palmitic Acid/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Apoptosis/physiology , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Humans , Hypoglycemic Agents/pharmacology , Mesenchymal Stem Cells/metabolism , Phosphorylation/drug effects , Ribonucleotides/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Clin Chim Acta ; 412(11-12): 970-4, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21300044

ABSTRACT

BACKGROUND: X-linked adrenoleukodystrophy (X-ALD) is a neurodegenerative disorder characterized by progressive demyelination of the nervous system, adrenocortical insufficiency and increase of very long chain fatty acids (VLCFAs) in the plasma and tissues. METHODS: A total of 131 individuals from 30 Chinese pedigrees were involved in this study, including 42 symptomatic patients, 44 female carriers, and 15 high-risk fetuses from 13 families. The mutation was first pinpointed through long distance RT-PCR-based RNA approach and confirmed through peripheral blood DNA approach. RESULTS: A total of 28 mutations were identified, of which 19 were missense, 3 nonsense and 6 frame-shift mutations. Thirteen mutations were novel, i.e. p.R280L, p.P580L, p.G343V, p.S108X, p.R259W, p.P534R, p.fs A246, p.L576P, p.K602X, p.A314P, p.N148D, p.H283R, and p.fs R89. Two mutations occurred de novo, for they were not found in somatic cells of their parents. Three females from the same family developed AMN-like symptoms and they were heterozygous for the p.H283R mutation. Four asymptomatic boys were diagnosed as X-ALD patients and prenatal molecular diagnosis were provided for 13 X-ALD-stricken families. CONCLUSIONS: Our work extended the spectrum of mutations in X-ALD and benefited genetic counseling through reliable identification of heterozygous females and asymptomatic males.


Subject(s)
Adrenoleukodystrophy/genetics , Laboratories , Molecular Diagnostic Techniques/methods , Mutation , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Animals , China , DNA Mutational Analysis , Female , Humans , Male , Molecular Sequence Data , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...