Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Zhongguo Dang Dai Er Ke Za Zhi ; 25(2): 223-228, 2023 Feb 15.
Article in Zh | MEDLINE | ID: mdl-36854702

ABSTRACT

Phosphomannomutase 2 deficiency is the most common form of N-glycosylation disorders and is also known as phosphomannomutase 2-congenital disorder of glycosylation (PMM2-CDG). It is an autosomal recessive disease with multi-system involvements and is caused by mutations in the PMM2 gene (OMIM: 601785), with varying severities in individuals. At present, there is still no specific therapy for PMM2-CDG, and early identification, early diagnosis, and early treatment can effectively prolong the life span of pediatric patients. This article reviews the advances in the diagnosis and treatment of PMM2-CDG.


Subject(s)
Congenital Disorders of Glycosylation , Humans , Child , Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/therapy , Mutation
2.
Pharmazie ; 76(8): 372-378, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34412736

ABSTRACT

Drug resistance caused by the extreme genetic variability of zhe hepatitis C virus has rendered effective combinations of drugs indispensable in the treatment of chronic hepatitis C (CHC). Herein, we developed a fixed-dose combination (FDC) treatment containing the NS5B inhibitor sofosbuvir (SOF) and the NS5A inhibitor fopitasvir (FOP). Then the dissolution behavior of FOP in FOP/SOF FDC was improved by co-micronizing FOP with lactose. The enhanced dissolution rate of FOP in the FDC was in good agreement with the behavior of the FOP singledrug tablet. In addition, pharmacokinetic studies showed that both FOP and SOF in the FDC exhibited similar characteristics (area under the curve, Cmax, Tmax, and T1/2) as those of tablets containing FOP or SOF alone. These results revealed that the FOP/SOF FDC represents a potential therapeutic option for the treatment of CHC.


Subject(s)
Hepatitis C, Chronic , Sofosbuvir , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Humans , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use , Tablets
3.
Bioorg Med Chem Lett ; 29(19): 126577, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31421967

ABSTRACT

Bromodomain-containing protein 4 (BRD4), a member of the bromodomain and extra-terminal (BET) family, has been recognized as an attractive candidate target for the treatment targeting gene transcription in several types of cancers. In this study, two types of novel compounds were designed, synthesized and evaluated as BRD4 inhibitors. Therein, pyridone derivatives were more effective against BRD4 protein and human leukemia cell lines MV4-11. Among them, compounds 11d, 11e and 11f were the most potential ones with IC50 values of 0.55 µM, 0.86 µM and 0.80 µM against BRD4, and exhibited remarkable antiproliferative activities against MV4-11 cells with IC50 values of 0.19 µM, 0.32 µM and 0.12 µM, respectively. Moreover, in western blot assay, compound 11e induced down-regulation of C-Myc, which is a significant downstream gene of BRD4. Cell cycle analysis assay also showed that compound 11e could block MV4-11 cells at G0/G1 phase. Taken together, our results suggested that compound 11e and its derivatives were a class of novel structural potential BRD4 inhibitors and could serve as lead compounds for further exploration.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Drug Design , Isoxazoles/chemistry , Leukemia/drug therapy , Pyridones/chemistry , Transcription Factors/antagonists & inhibitors , Cell Cycle , Humans , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Tumor Cells, Cultured
4.
Exp Cell Res ; 340(2): 227-37, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26748182

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are capable of unlimited self-renewal and can generate nearly all cells in the body. Changes induced by different LSD1 activities on the regulation of hiPSC self-renewal and differentiation and the mechanism underlying such changes were determined. We used two different LSD1 inhibitors (phenelzine sulfate and tranylcypromine) and RNAi technique to inhibit LSD1 activity, and we obtained hiPSCs showing 71.3%, 53.28%, and 31.33% of the LSD1 activity in normal hiPSCs. The cells still maintained satisfactory self-renewal capacity when LSD1 activity was at 71.3%. The growth rate of hiPSCs decreased and cells differentiated when LSD1 activity was at approximately 53.28%. The hiPSCs were mainly arrested in the G0/G1 phase and simultaneously differentiated into endodermal tissue when LSD1 activity was at 31.33%. Teratoma experiments showed that the downregulation of LSD1 resulted in low teratoma volume. When LSD1 activity was below 50%, pluripotency of hiPSCs was impaired, and the teratomas mainly comprised endodermal and mesodermal tissues. This phenomenon was achieved by regulating the critical balance between histone methylation and demethylation at regulatory regions of several key pluripotent and developmental genes.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Self Renewal , Histone Demethylases/metabolism , Induced Pluripotent Stem Cells/cytology , Cell Line , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Histones/metabolism , Humans , Methylation , Phenelzine/pharmacology , RNA Interference , Tranylcypromine/pharmacology
5.
Mol Cancer ; 14: 56, 2015 Mar 07.
Article in English | MEDLINE | ID: mdl-25890196

ABSTRACT

BACKGROUND: Acute myeloid leukemia (AML) is initiated and maintained by a subset of self-renewing leukemia stem cells (LSCs), which contribute to the progression, recurrence and therapeutic resistance of leukemia. However, the mechanisms underlying the maintenance of LSCs drug resistance have not been fully defined. In this study, we attempted to elucidate the mechanisms of LSCs drug resistance. METHODS: We performed reverse phase protein arrays to analyze the expression of anti-apoptotic proteins in the LSC-enriched leukemia cell line KG-1a. Immuno-blotting, cell viability and clinical AML samples were evaluated to verify the micro-assay results. The characteristics and transcriptional regulation of survivin were analyzed with the relative luciferase reporter assay, mutant constructs, chromatin immuno-precipitation (ChIP), quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), and western blotting. The levels of Sp1, c-Myc, phospho-extracellular signal-regulated kinase (p-ERK), phospho-mitogen and stress-activated protein kinase (p-MSK) were investigated in paired CD34+ and CD34- AML patient samples. RESULTS: Survivin was highly over-expressed in CD34 + CD38- KG-1a cells and paired CD34+ AML patients compared with their differentiated counterparts. Functionally, survivin contributes to the drug resistance of LSCs, and Sp1 and c-Myc concurrently regulate levels of survivin transcription. Clinically, Sp1 and c-Myc were significantly up-regulated and positively correlated with survivin in CD34+ AML patients. Moreover, Sp1 and c-Myc were further activated by the ERK/MSK mitogen-activated protein kinase (MAPK) signaling pathway, modulating survivin levels. CONCLUSION: Our findings demonstrated that ERK/MSK/Sp1/c-Myc axis functioned as a critical regulator of survivin expression in LSCs, offering a potential new therapeutic strategy for LSCs therapy.


Subject(s)
Drug Resistance, Neoplasm/genetics , Inhibitor of Apoptosis Proteins/genetics , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Sp1 Transcription Factor/genetics , Adult , Antigens, CD34/genetics , Cell Line, Tumor , Cell Survival/genetics , Female , HL-60 Cells , Humans , K562 Cells , Male , Neoplastic Stem Cells/metabolism , Signal Transduction/genetics , Survivin , Transcription, Genetic/genetics , U937 Cells , Up-Regulation/genetics
6.
Stem Cell Res Ther ; 11(1): 185, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32430053

ABSTRACT

BACKGROUND: Human induced pluripotent stem cells (hiPSCs) represent a potentially unlimited source of pancreatic endocrine lineage cells. Although insulin-producing ß cells derived from hiPSCs have been successfully induced, much work remains to be done to achieve mature ß cells. Lysine-specific demethylase 1 (LSD1) plays an important role in the regulation of hiPSC self-renewal and differentiation. We propose a new strategy to acquire insulin-producing cells (IPCs) from hiPSCs by knocking down LSD1. METHODS: Knockdown of LSD1 in hiPSCs with five shRNA. Assessment of the effects of shRNA on hiPSC proliferation, cell cycle, and apoptosis. Using knockdown hiPSCs with 31.33% LSD1 activity, we achieved a four-step differentiation into IPCs and test its differentiation efficiency, morphology, and marker genes and proteins. We implanted the IPCs into the renal subcapsular of SCID-Beige diabetic mice to evaluate the hypoglycemic effect in vivo. We tested LSD1 and HDAC1 whether they are present in the CoREST complex through IP-WB, and analyzed LSD1, CoREST, HDAC1, H3K4me2/me3, and H3K27me3 protein expression before and after knockdown of LSD1. RESULTS: Differentiated hiPSCs were 38.32% ± 3.54% insulin-positive cells and released insulin/C-peptide in response to glucose stimulus in a manner comparable to adult human islets. Most of the IPCs co-expressed mature ß cell-specific markers. When transplanted under the left renal capsule of SCID-Beige diabetic mice, these IPCs reversed hyperglycemia, leading to a significant increase in the definitive endoderm cells. IP-WB results showed that LSD1, HDAC1, and CoREST formed a complex in hiPSCs. Chip-PCR results showed that LSD1, HDAC1, and CoREST were enriched in the same district during the SOX17 and FOXA2 promoter region. Inhibition of LSD1 would not affect the level of CoREST but decreased the HDAC1 expressions. The H3K4me2/me3 and H3K9act level of SOX17 and FOXA2 promoter region increased after inhibited of LSD1, and promoted transcriptional activation. The H3K4me2/me3 and H3K9act level of OCT4 and SOX2 promoter region decreased with the transcriptional repressed. CONCLUSIONS: LSD1 regulated histone methylation and acetylation in promoter regions of pluripotent or endodermal genes. Our results suggest a highly efficient approach to producing IPCs from hiPSCs.


Subject(s)
Diabetes Mellitus, Experimental , Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Animals , Cell Differentiation , Histone Demethylases/genetics , Humans , Insulin , Mice , Mice, SCID
7.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 25(2): 572-576, 2017 Apr.
Article in Zh | MEDLINE | ID: mdl-28446313

ABSTRACT

OBJECTIVE: To investigate the effects of Agkistrodon acutus venom protein C activator(PCA) on ultrastructure of human umbilical vein endothelial cells(HUVEC), and the levels of tissue factor(TF), vascular von Willebrand factor (vWF) and endothelin-1 secreted by HUVEC and to clarify the anti-thrombotic mechanism of PCA. METHODS: The experiments were divided into control group(DMEM), LPS group (LPS 0.1 µg/ml), PCA group(PCA 1 µg/ml) and PCA+LPS group (1 µg/ml PCA+ 0.1 µg/ml LPS). The morphology of endoplasmic reticulum, mitochondria and the number of autophagosome in HUVEC were observed by transmission electron microscopy. The TF, vWF and ET-1 were measured in the medium of each group by ELISA; RT-PCR was used to detect mRNA expression level of vWF and ET-1 in cells; and the protein expression level of TF in cells was detected by Western blot. RESULTS: Compared with the control group, the ultrastructural changes of HUVEC in the LPS group included the cell membrane getting rough, swelling of mitochondria and endoplasmic reticulum, and autophagosome increase, however, the ultrastructure differences between PCA and control group were not significant. Compared with the ultrastructure of HUVECs in LPS group, the swelling of mitochondria and endoplasmic reticulum disappeared in the LPS+PCA group, and the number of autophagosome decreased obviously. Compared with the control group, the content of ET-1, vWF and TF in cell culture supernatant, and the protein expression level of vWF, ET-1 gene and TF protein were significantly increased in LPS group (P<0.05); the expression levels of the 3 factors in the cell culture supernatant and cells in PCA group were not significantly different from the control group (P>0.05). The expression levels of TF, vWF and ET-1 in LPS group were significantly lower than those in LPS+PCA group (P<0.05). CONCLUSION: PCA(1 µg/ml) can reduce the ultrastructural changes of HUVEC induced by LPS, and inhibit the increase of TF, vWF and ET-1 secretion from HUVEC induced by LPS.


Subject(s)
Agkistrodon , Human Umbilical Vein Endothelial Cells/drug effects , Snake Venoms/pharmacology , Animals , Cells, Cultured , Endothelium, Vascular , Humans , Thromboplastin/drug effects , Thromboplastin/metabolism , Venoms , von Willebrand Factor
8.
Sci Rep ; 6: 19995, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26847520

ABSTRACT

Drug resistance is one of the leading causes of failed cancer therapy in the treatment of acute myeloid leukemia. Although the mechanisms of resistance are poorly understood, they may be related to the presence of leukemia stem cells (LSCs). Down-regulation of the miR-203 reportedly contributes to oncogenesis and chemo-resistance in multiple cancers. We found that miR-203 expression was down-regulated in CD34 + AML cells as compared with CD34- cells isolated from patients as well as in LSC-enriched (CD34 + CD38-) cell lines KG-1a or MOLM13. Additionally, re-expression of miR-203 led to decreased cell proliferation, self-renewal, and sphere formation in LSCs. Moreover, miR-203 was found to directly target the 3'un-translated regions of survivin and Bmi-1 mRNAs affecting proliferation and self-renewal in LSCs. In this study, we identified a novel miR-203/survivin/Bmi-1 axis involved in the regulation of biological properties of LSCs. This axis may represent a new therapeutic target for acute myeloid leukemia and a potential prognosis/diagnostic marker for LSCs therapy.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , 3' Untranslated Regions , Animals , Antigens, CD34/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/therapeutic use , Neoplastic Stem Cells/cytology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Sequence Alignment , Survivin
9.
PLoS One ; 9(4): e94548, 2014.
Article in English | MEDLINE | ID: mdl-24722487

ABSTRACT

Islet transplantation has considerable potential as a cure for diabetes. However, the difficulties that arise from inflammation and the immunological rejection of transplants must be addressed for islet transplantation to be successful. Alpha 1-antitrypsin (AAT) inhibits the damage on ß cells caused by inflammatory reactions and promotes ß-cell survival and proliferation. This protein also induces specific immune tolerance to transplanted ß cells. However, whether the expression of AAT in ß cells themselves could eliminate or decrease immunological rejection of transplants is not clear. Therefore, we established a ß cell line (NIT-hAAT) that stably expresses human AAT. Interestingly, in a cytotoxic T lymphocyte (CTL)-killing assay, we found that hAAT reduced apoptosis and inflammatory cytokine production in NIT-1 cells and regulated the Th1/Th2 cytokine balance in vitro. In vivo transplantation of NIT-hAAT cells into mice with diabetes showed hAAT inhibited immunological rejection for a short period of time and increased the survival of transplanted ß cells. This study demonstrated that hAAT generated remarkable immunoprotective and immunoregulation effects in a model of ß cell islet transplantation for diabetes model.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Graft Rejection/immunology , Immunologic Factors/genetics , Immunomodulation/genetics , Insulin-Secreting Cells/transplantation , alpha 1-Antitrypsin/genetics , Animals , Apoptosis , Cell Survival , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression , Graft Rejection/prevention & control , Humans , Immune Tolerance , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Th1-Th2 Balance , Transgenes , alpha 1-Antitrypsin/immunology
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 28(2): 120-3, 2012 Feb.
Article in Zh | MEDLINE | ID: mdl-22304765

ABSTRACT

AIM: To construct the prokaryotic expression plasmid pGEX-4T-1-BZLF1N-BLRF2, and express it in Escherichia coli. METHODS: The EB virus BZLF1N gene and BLRF2 gene were amplified by RT-PCR respectively. Then, the two genes were linked by splicing overlap extension PCR method and inserted into the vector pGEX-4T-1, and the recombinant plasmid pGEX-4T-1-BZLF1N-BLRF2 was transformed into E.coli BL21 (DE3) strain. The expression protein ZtaN-p23 was analysed by SDS-PAGE and immunoreactivity was proved by Western blotting. RESULTS: Restriction enzyme digestion and DNA sequencing showed recombinant plasmid constructed successfully. The expression product ZtaN-p23 with the molecular weight 46000 was located in the cytoplasm and insoluble. The ZtaN-p23 up to 95% purity was obtained after purified using affinity chromatography. Western blotting showed fusion protein possessed a well bioactivity and specificity. CONCLUSION: The fusion gene BZLF1N-BLRF2 is successfully constructed and effectively expressed in E.coli, which lay the foundation for further research on its biological properties and functions.


Subject(s)
Escherichia coli/genetics , Gene Expression , Herpesvirus 4, Human/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Trans-Activators/genetics , Trans-Activators/immunology , Escherichia coli/metabolism , Herpesvirus 4, Human/immunology , Humans , Molecular Weight , Recombinant Fusion Proteins/isolation & purification , Solubility
11.
J Physiol Biochem ; 68(4): 603-10, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22644623

ABSTRACT

To maintain islets survival and function is critical in successful pancreatic transplantation. Pancreatic progenitors cells (PPCs) with lineage potentials, giving rise to exocrine, endocrine, and duct cells, reside in developing and adult pancreas. As tissue-specific stem cells, they can produce pancreatic tissue-specific matrix factors to promote islets survival and function. The aim of our research was to investigate the protective effect of rat pancreatic-duodenal homeobox 1 (Pdx1)(+)/nestin(+) PPCs on islets. In vitro, co-culturing islets with Pdx1(+)/nestin(+) PPCs prolonged the former survival from 7 to 14 days. Furthermore, with high glucose (300.8 mg/dl) stimuli, the yield of insulin in co-cultures was significantly higher than that in control group (single islets group). In vivo, co-transplanting islets and Pdx1(+)/nestin(+) PPCs for 3 days, the blood glucose of diabetic rat was significantly decreased to normal level and sustained for 2 weeks. Without Pdx1(+)/nestin(+) PPCs in islets transplantation, hyperglycemia was reversed at day 7 and recovered at day 15. Pathology analysis showed that islets had remnants in co-transplantation at day 21, as complete graft rejection in alone islets transplantation. Our study showed that Pdx1(+)/nestin(+) PPCs displayed the ability of preserving islets viability and function in vitro and prolonging their survival in vivo.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Homeodomain Proteins/metabolism , Islets of Langerhans/physiopathology , Pancreas/pathology , Stem Cell Transplantation , Stem Cells/metabolism , Trans-Activators/metabolism , Animals , Blood Glucose , Cell Survival , Cells, Cultured , Coculture Techniques , Female , Insulin/metabolism , Insulin Secretion , Intermediate Filament Proteins/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Kidney/pathology , Nerve Tissue Proteins/metabolism , Nestin , Pregnancy , Rats , Rats, Sprague-Dawley
12.
J Exp Clin Cancer Res ; 30: 98, 2011 Oct 17.
Article in English | MEDLINE | ID: mdl-21999220

ABSTRACT

BACKGROUND: DNA methylation has been widely used in classification, early diagnosis, therapy and prediction of metastasis as well as recurrence of cervical cancer. DNMT methyltransferase 1 (DNMT1), which plays a significant role in maintaining DNA methylation status and regulating the expression of tumor suppressor genes. The aim of this research was to investigate the relationship between DNMT1 and abnormal methylation of tumor suppressor genes and malignant phenotype in cervical cancer. METHODS: Levels of DNMT1 mRNA and protein were detected using qPCR and Western blot, respectively. Cell proliferation was analyzed by MTT and apoptosis was performed by Annexin V-FITC/PI double staining flow cytometry, respectively. MeDIP-qPCR and qPCR were performed to measure demethylation status and mRNA re-expression level of 7 tumor-suppressor genes (CCNA1, CHFR, FHIT, PAX1, PTEN, SFRP4, TSLC1) in Hela and Siha cells after silencing DNMT1. RESULTS: The average expression levels of DNMT1 mRNA and protein in Hela and Siha cells were decreased significantly compared with control group. The flow cytometry and MTT results showed that Hela and Siha cells apoptosis rates and cell viabilities were 19.4 ± 2.90%, 25.7 ± 3.92% as well as 86.7 ± 3.12%, 84.16 ± 2.67% respectively 48 h after transfection (P < 0.01). Furthermore, the promoter methylation of five tumor suppressor genes was decreased with the increased mRNA expression after silencing DNMT1, whereas there were no significant changes in PTEN and FHIT genes in Hela cells, and CHFR and FHIT genes in Siha cells. CONCLUSIONS: Our experimental results demonstrate that methylation status of DNMT1 can influence several important tumor suppressor genes activity in cervical tumorigenesis and may have the potential to become an effective target for treatment of cervical cancer.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Gene Silencing , Genes, Tumor Suppressor/physiology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Apoptosis , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cell Proliferation , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA, Neoplasm/genetics , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Phenotype , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Uterine Cervical Neoplasms/metabolism
13.
J. physiol. biochem ; 68(4): 603-610, dic. 2012.
Article in English | IBECS (Spain) | ID: ibc-122308

ABSTRACT

To maintain islets survival and function is critical in successful pancreatic transplantation. Pancreatic progenitors cells (PPCs) with lineage potentials, giving rise to exocrine, endocrine, and duct cells, reside in developing and adult pancreas. As tissue-specific stem cells, they can produce pancreatic tissue-specific matrix factors to promote islets survival and function. The aim of our research was to investigate the protective effect of rat pancreatic-duodenal homeobox 1 (Pdx1)+/nestin+ PPCs on islets. In vitro, co-culturing islets with Pdx1+/nestin+ PPCs prolonged the former survival from 7 to 14 days. Furthermore, with high glucose (300.8 mg/dl) stimuli, the yield of insulin in co-cultures was significantly higher than that in control group (single islets group). In vivo, co-transplanting islets and Pdx1+/nestin+ PPCs for 3 days, the blood glucose of diabetic rat was significantly decreased to normal level and sustained for 2 weeks. Without Pdx1+/nestin+ PPCs in islets transplantation, hyperglycemia was reversed at day 7 and recovered at day 15. Pathology analysis showed that islets had remnants in co-transplantation at day 21, as complete graft rejection in alone islets transplantation. Our study showed that Pdx1+/nestin+ PPCs displayed the ability of preserving islets viability and function in vitro and prolonging their survival in vivo (AU)


Subject(s)
Animals , Rats , Stem Cells , Islets of Langerhans , Pancreas Transplantation , Protective Agents/pharmacokinetics , Disease Models, Animal , Case-Control Studies
SELECTION OF CITATIONS
SEARCH DETAIL