Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Clin Endocrinol Diabetes ; 128(2): 89-96, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30641609

RESUMO

Demyelination resulting from Schwann cell injury is a main pathological feature of diabetic neuropathy, and a key contributor to this process may be inflammation due to advanced glycation end products (AGEs). Therefore, protection by anti-inflammation agents is anticipated. In this study, we showed that interleukin-10 (IL-10), an anti-inflammatory cytokine, inhibits apoptosis of Schwann cells induced by AGEs in vitro. We isolated and cultured Schwann cells from rat sciatic nerves. As detected by flow cytometry, apoptosis of Schwann cells markedly increased following incubation with AGEs for 48 h. However, pretreatment with IL-10 inhibited AGE-induced apoptosis. The effect of IL-10 on NF-κB, which is a very important regulator of inflammation, was also evaluated, and results showed high levels of phospho-NF-κB and nuclear localization of NF-κB in cells incubated with AGEs but low levels of phospho-NF-κB and cytoplasmic localization in the cells incubated with IL-10, indicating the activation of NF-κB by AGEs and inhibition of NF-κB by IL-10. Moreover, incubating Schwann cells with an NF-κB inhibitor (caffeic acid phenethyl ester) for 30 min before adding AGEs mimicked IL-10, lowering the amount of reactive oxygen species and activity of caspase-3 and also decreasing apoptosis in Schwann cells. These results indicate that IL-10 may protect Schwann cells against AGE-induced apoptosis by attenuating oxidative stress via the inhibition of activation of NF-κB.


Assuntos
Apoptose/fisiologia , Neuropatias Diabéticas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Interleucina-10/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células de Schwann/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Masculino , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ratos , Ratos Wistar , Células de Schwann/efeitos dos fármacos , Nervo Isquiático/citologia , Quinase Induzida por NF-kappaB
2.
Life Sci ; 225: 107-116, 2019 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-30928409

RESUMO

AIMS: To explore the effect and mechanism of 1, 25-(OH)2D3 on Schwann cell apoptosis induced by advanced glycation end products. MAIN METHODS: Schwann cells, isolated from rodent sciatic nerve were incubated with AGE-modified bovine serum albumin(AGE) to mimic diabetic conditions and 1,25-(OH)2D3 was used as protector. Cell apoptosis was detected by PI/Annexin-V staining, caspase 3 activity assay and western blotting for caspase 3 and PARP. The activation of protein kinase A (PKA) and nuclear factor kappa-B (NF-κB) was evaluated by western blot. Immunofluorescent staining was used for intercellular location of NF-κB. Cytokine secretion was evaluated by enzyme-linked immunosorbent assay. KEY FINDINGS: Schwann cell apoptosis accelerated after incubating with AGE. However, if combining 1,25-(OH)2D3 with AGE, apoptosis decreased significantly. 1,25-(OH)2D3 enhanced PKA activity, but inhibited AGE-induced nuclear translocation of NF-κB. Furthermore, PKA activator (8-bromoadenoside cyclic adenoside monophosphate, 8-Br-cAMP) or NF-κB inhibitor (caffeic acid phenethyl ester, CAPE) could reduce the apoptosis, decreased cleaved caspase 3 and cleaved PARP, suggesting the involvement of PKA and NF-κB pathways in the protection of 1,25-(OH)2D3 on Schwann cells. Moreover, 8-Br-cAMP and CAPE could inhibit AGE-induced secretion of interleukin(IL)-1ß, prostaglandin E2(PEG2) and cyclooxygenase 2(COX2). Interestingly, 8-Br-cAMP decreased phospho-NF-κB and inhibited nucleus translocation of NF-κB. It hinted at the regulation of PKA to NF-κB. Finally, a pre-treatment of H-89 (an inhibitor of PKA) could block the protection of 1,25-(OH)2D3 on cell apoptosis. In conclusion, 1,25-(OH)2D3 could protect Schwann cell against AGE-induced apoptosis through PKA/NF-κB pathway. SIGNIFICANCE: These findings provide experimental rationales for using vitamin D for diabetic neuropathy.


Assuntos
Apoptose/efeitos dos fármacos , Calcitriol/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Produtos Finais de Glicação Avançada/farmacologia , NF-kappa B/metabolismo , Substâncias Protetoras/farmacologia , Células de Schwann/efeitos dos fármacos , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/genética , Regulação da Expressão Gênica/efeitos dos fármacos , NF-kappa B/genética , Ratos , Ratos Wistar , Células de Schwann/metabolismo , Células de Schwann/patologia , Transdução de Sinais , Vitaminas/farmacologia
3.
J Diabetes Investig ; 10(3): 613-625, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30307132

RESUMO

AIMS/INTRODUCTION: Blockade or reversal the progression of diabetic nephropathy is a clinical challenge. The aim of the present study was to examine whether recombinant human glucagon-like peptide-1 (rhGLP-1) has an effect on alleviating urinary protein and urinary albumin levels in diabetic rats. MATERIALS AND METHODS: Streptozotocin-induced diabetes rats were treated with rhGLP-1 insulin and saline. Using immunostaining, hematoxylin-eosin, electron microscopy and periodic acid-Schiff staining to study the pathology of diabetic nephropathy, and we carried out quantitative reverse transcription polymerase chain reaction, western blot and immunohistochemistry to identify the differentially expressed proteins. The mechanism was studied through advanced glycation end-products-induced tubular epithelial cells. RESULTS: rhGLP-1 inhibits protein kinase C (PKC)-ß, but increases protein kinase A (PKA), which reduces oxidative stress in glomeruli and in cultured glomerular microvascular endothelial cells. In tubules, rhGLP-1 increased the expression of two key proteins related to re-absorption - megalin and cubilin - which was accompanied by downregulation of PKC-ß and upregulation of PKA. On human proximal tubular epithelial cells, rhGLP-1 enhanced the absorption of albumin, and this was blocked by a PKC activator or PKA inhibitor. CONCLUSIONS: These findings suggest that rhGLP-1 can reverse diabetic nephropathy by protecting both glomeruli and tubules by inhibiting PKC and activating PKA.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/prevenção & controle , Peptídeo 1 Semelhante ao Glucagon/administração & dosagem , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteínas Recombinantes/administração & dosagem , Animais , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/etiologia , Feminino , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Masculino , Estresse Oxidativo , Ratos , Ratos Wistar
4.
Diabetes ; 67(7): 1345-1355, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29716892

RESUMO

We investigated how human proislet peptide (HIP) regulates differentiation of human fetus-derived pancreatic progenitor cells (HFPPCs) and explored the potential link between HIP signaling and the menin pathway, which is key to regulating pancreatic islet differentiation. The data show that HIP promoted expression of proislet transcription factors (TFs), including PDX-1, MAFA, and NKX6.1, as well as other maturation markers of ß-cells, such as insulin, GLUT2, KIR6.2, SUR1, and VDCC. Moreover, HIP increased insulin content and promoted the ability of HFPPCs to normalize blood glucose in diabetic mice. HIP inhibited the TF FOXO1 by increasing AKT-mediated phosphorylation. HIP-induced repression of FOXO1 suppressed menin expression, leading to reducing menin binding to the promoter of the three key proislet TFs, decreasing recruitment of H3K9 methyltransferase SUV39H1, and thus reducing repressive H3K9me3 at the promoter. These coordinated actions lead to increased expression of the proislet TFs, resulting in induction of HFPPC differentiation. Consistently, constitutive activation of FOXO1 blocks HIP-induced transcription of these TFs. Together, these studies unravel the crucial role of the HIP/AKT/FOXO/menin axis in epigenetically controlling expression of proislet TFs, regulating the differentiation of HFPPCs, and normalizing blood glucose in diabetic mice.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/genética , Epigênese Genética/efeitos dos fármacos , Proteína Forkhead Box O1/genética , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas/genética , Células-Tronco/efeitos dos fármacos , Animais , Diferenciação Celular/genética , Células Cultivadas , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/terapia , Células HEK293 , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/transplante , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células-Tronco/fisiologia
5.
Oncol Lett ; 15(4): 4255-4261, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29541192

RESUMO

Aberrant blood vessel formation and hemorrhage may contribute to tumor progression and are potential targets in the treatment of several types of cancer. Pancreatic neuroendocrine tumors (PNETs) are highly vascularized, particularly when they are well-differentiated. However, the process of vascularization and endothelial cell detachment in PNETs is poorly understood. In the present study, 132 PNET clinical samples were examined and a special type of hemorrhagic region was observed in ~30% of the samples regardless of tumor subtype. These hemorrhagic regions were presented as blood-filled caverns with a smooth boundary and were unlined by endothelial cells. Based on the extensive endothelial cell detachment observed in the clinical samples, the formation process of these blood-filled caverns was hypothesized. Blood vessel dilation followed by detachment of endothelial cells from the surrounding tumor tissue was speculated. This was further supported using an INS-1 xenograft insulinoma model. As the formation process was distinct from the typical diffusive hemorrhage, it was named 'pseudo-hemorrhage'. Furthermore, it was demonstrated that epithelial (E-) cadherin and ß-catenin were overexpressed in tumor cells surrounding these pseudo-hemorrhagic regions. Therefore, even though no statistically significant association of pseudo-hemorrhage with clinical features (metastasis or disease recurrence) was identified, the high levels of E-cadherin and ß-catenin expression may suggest that a number of features of normal islet cells are retained.

6.
Stem Cell Res Ther ; 9(1): 27, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29402326

RESUMO

BACKGROUND: The study aimed to evaluate the efficacy and safety of gingival mesenchymal stem cells (GMSCs) from human fetal gingival tissue used for treating gingival defects in a rat model. METHODS: GMSCs were isolated from human fetal gingival tissue and identified by flow cytometry for nestin, Oct4, vimentin, NANOG, CD105, and CD90. The immunogenicity of GMSCs was analyzed by mixed lymphocyte reactions; the tumorigenicity of GMSCs was evaluated by xenotransplanting into nude mice. The gingival defect animal model was established by mechanical resection in rats. GMSCs were transplanted into the defective area, and the regeneration of gingival tissue was observed twice weekly. Four weeks after transplantation, the gingival tissue was surgically cut down, and the graft was analyzed by immunohistochemistry staining for human mitochondrial antigens and rat CD3 and CD20. RESULTS: GMSCs from human fetal gingival tissue positively expressed nestin, Oct4, vimentin, NANOG, CD105, and CD90. There was no cell aggregation after mixed lymphocyte reactions, and interleukin-2 did not increase. Inoculation of GMSCs into nude mice for 6 months showed no tumor formation. GMSCs were transplanted into the gingiva defects of rats. One week after transplantation, the defect area was reduced, and after 3 weeks the morphology and color of local gingival tissue was similar to normal gingival tissue, and gingival height was the same as the normal control group. CONCLUSIONS: Using GMSCs from human fetal gingival tissue to treat gingival defects is a safe and effective innovative treatment method.


Assuntos
Antígenos de Diferenciação/biossíntese , Feto , Gengiva , Doenças da Gengiva , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Gengiva/lesões , Gengiva/metabolismo , Gengiva/patologia , Doenças da Gengiva/metabolismo , Doenças da Gengiva/patologia , Doenças da Gengiva/terapia , Xenoenxertos , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Ratos , Ratos Wistar
7.
Biochem Biophys Res Commun ; 495(1): 793-800, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29137984

RESUMO

GLP-1-based treatment improves glycemia through stimulation of insulin secretion and inhibition of glucagon secretion. Recently, more and more findings showed that GLP-1 could also protect kidney from diabetic nephropathy. Most of these studies focused on glomeruli, but the effect of GLP-1 on tubulointerstitial and tubule is not clear yet. In this study, we examined the renoprotective effect of recombinant human GLP-1 (rhGLP-1), and investigated the influence of GLP-1 on inflammation and tubulointerstitial injury using diabetic nephropathy rats model of STZ-induced. The results showed that rhGLP-1 reduced urinary albumin without influencing the body weight and food intake. rhGLP-1 could increased the serum C-peptide slightly but not lower fasting blood glucose significantly. In diabetic nephropathy rats, beside glomerular sclerosis, tubulointerstitial fibrosis was very serious. These lesions could be alleviated by rhGLP-1. rhGLP-1 decreased the expression of profibrotic factors collagen I, α-SMA, fibronectin, and inflammation factors MCP-1 and TNFα in tubular tissue and human proximal tubular cells (HK-2 cells). Furthermore, rhGLP-1 significantly inhibited the phosphorylation of NF-κB, MAPK in both diabetic tubular tissue and HK-2 cells. The inhibition of the expression of TNFα, MCP-1, collagen I and α-SMA in HK-2 cells by GLP-1 could be mimicked by blocking NF-κB or MAPK. These results indicate that rhGLP-1 exhibit renoprotective effect by alleviation of tubulointerstitial injury via inhibiting phosphorylation of MAPK and NF-κB. Therefore, rhGLP-1 may be a potential drug for treatment of diabetic nephropathy.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Nefrite Intersticial/tratamento farmacológico , Nefrite Intersticial/metabolismo , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Peptídeo 1 Semelhante ao Glucagon/genética , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , NF-kappa B/metabolismo , Nefrite Intersticial/patologia , Ratos , Proteínas Recombinantes/uso terapêutico , Resultado do Tratamento
8.
J Transl Med ; 15(1): 147, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28655312

RESUMO

BACKGROUND: Diabetic nephropathy (DN) is a severe complication of diabetes mellitus (DM). Pancreas or islet transplantation has been reported to prevent the development of DN lesions and ameliorate or reverse existing glomerular lesions in animal models. Shortage of pancreas donor is a severe problem. Islets derived from stem cells may offer a potential solution to this problem. OBJECTIVE: To evaluate the effect of stem cell-derived islet transplantation on DN in a rat model of streptozotocin-induced DM. METHODS: Pancreatic progenitor cells were isolated from aborted fetuses of 8 weeks of gestation. And islets were prepared by suspension culture after a differentiation of progenitor cells in medium containing glucagon-like peptide-1 (Glp-1) and nicotinamide. Then islets were transplanted into the liver of diabetic rats via portal vein. Blood glucose, urinary volume, 24 h urinary protein and urinary albumin were measured once biweekly for 16 weeks. Graft survival was evaluated by monitoring human C-peptide level in rat sera and by immunohistochemical staining for human mitochondrial antigen and human C-peptide in liver tissue. The effect of progenitor-derived islets on filtration membrane was examined by electron microscopy and real-time polymerase chain reaction (PCR). Immunohistochemical staining, real-time PCR and western blot were employed for detecting fibronectin, protein kinase C beta (PKCß), protein kinase A (PKA), inducible nitric oxide synthase (iNOS) and superoxide dismutase (SOD). RESULTS: Islet-like clusters derived from 8th gestational-week human fetal pancreatic progenitors survived in rat liver. And elevated serum level of human C-peptide was detected. Blood glucose, 24 h urinary protein and urinary albumin were lower in progenitor cell group than those in DN or insulin treatment group. Glomerular basement membrane thickness and fibronectin accumulation decreased significantly while podocytes improved morphologically in progenitor cell group. Furthermore, receptor of advanced glycation end products and PKCß became down-regulated whereas PKA up-regulated by progenitor cell-derived islets. And iNOS rose while SOD declined. CONCLUSIONS: DN may be reversed by transplantation of human fetal pancreatic progenitor cell-derived islets. And fetal pancreatic progenitor cells offer potential resources for cell replacement therapy.


Assuntos
Nefropatias Diabéticas/terapia , Feto/citologia , Rim/lesões , Pâncreas/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Albuminúria/sangue , Albuminúria/complicações , Albuminúria/patologia , Animais , Glicemia/metabolismo , Sobrevivência Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/complicações , Nefropatias Diabéticas/fisiopatologia , Taxa de Filtração Glomerular , Humanos , Rim/patologia , Rim/fisiopatologia , Rim/ultraestrutura , Fígado/patologia , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo , Proteína Quinase C beta/metabolismo , Ratos Wistar , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Estreptozocina , Superóxido Dismutase/metabolismo
9.
Int J Mol Med ; 40(1): 130-136, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28534945

RESUMO

Advanced glycation end products (AGEs), which accumulate in the body during the development of diabetes, may be one of the factors leading to pancreatic ß-cell failure and reduced ß-cell mass. However, the mechanisms responsible for AGE­induced apoptosis remain unclear. This study identified the role and mechanisms of action of tribbles homolog 3 (TRB3) in AGE-induced ß-cell oxidative damage and apoptosis. Rat insulinoma cells (INS-1) were treated with 200 µg/ml AGEs for 48 h, and cell apoptosis was then detected by TUNEL staining and flow cytometry. The level of intracellular reactive oxygen species (ROS) was measured by a fluorescence assay. The expression levels of receptor of AGEs (RAGE), TRB3, protein kinase C ß2 (PKCß2) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) were evaluated by RT-qPCR and western blot analysis. siRNA was used to knockdown TRB3 expression through lipofection, followed by an analysis of the effects of TRB3 on PKCß2 and NOX4. Furthermore, the PKCß2-specific inhibitor, LY333531, was used to analyze the effects of PKCß2 on ROS levels and apoptosis. We found that AGEs induced the apoptosis of INS-1 cells and upregulated RAGE and TRB3 expression. AGEs also increased ROS levels in ß-cells. Following the knockdown of TRB3, the AGE-induced apoptosis and intracellular ROS levels were significantly decreased, suggesting that TRB3 mediated AGE-induced apoptosis. Further experiments demonstrated that the knockdown of TRB3 decreased the PKCß2 and NOX4 expression levels. When TRB3 was knocked down, the cells expressed decreased levels of PKCß2 and NOX4. The PKCß2­specific inhibitor, LY333531, also reduced AGE-induced apoptosis and intracellular ROS levels. Taken together, our data suggest that TRB3 mediates AGE-induced oxidative injury in ß-cells through the PKCß2 pathway.


Assuntos
Apoptose , Produtos Finais de Glicação Avançada/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Quinase C beta/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Produtos Finais de Glicação Avançada/genética , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo , Proteína Quinase C beta/antagonistas & inibidores , Proteína Quinase C beta/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos
10.
J Diabetes Res ; 2017: 6726901, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28349071

RESUMO

Our colleagues have reported previously that human pancreatic progenitor cells can readily differentiate into insulin-containing cells. Particularly, transplantation of these cell clusters upon in vitro induction for 3-4 w partially restores hyperglycemia in diabetic nude mice. In this study, we used human fetal pancreatic progenitor cells to identify the forkhead protein FoxO1 as the key regulator for cell differentiation. Thus, induction of human fetal pancreatic progenitor cells for 1 week led to increase of the pancreatic ß cell markers such as Ngn3, but decrease of stem cell markers including Oct4, Nanog, and CK19. Of note, FoxO1 knockdown or FoxO1 inhibitor significantly upregulated Ngn3 and insulin as well as the markers such as Glut2, Kir6.2, SUR1, and VDCC, which are designated for mature ß cells. On the contrary, overexpression of FoxO1 suppressed the induction and reduced expression of these ß cell markers. Taken together, these results suggest that FoxO1 may act as a repressor to inhibit cell differentiation in human fetal pancreatic progenitor cells.


Assuntos
Diferenciação Celular/fisiologia , Proteína Forkhead Box O1/metabolismo , Pâncreas/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Células Cultivadas , Proteína Forkhead Box O1/genética , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Pâncreas/citologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Células-Tronco , Receptores de Sulfonilureias/genética , Receptores de Sulfonilureias/metabolismo , Regulação para Cima
11.
Am J Cancer Res ; 6(10): 2277-2288, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27822417

RESUMO

Tumor endothelial cells have been found to be associated with metastasis and cancer progression. In this study, we reported that human esophageal cancer endothelial cells (HECEC), unlike corresponding human esophageal normal endothelial cells (HENEC) displayed several distinct feature couple with unique gene expression profile. Further studies showed that HECEC can enhance migration, invasion and self-renewal properties of esophageal carcinoma cell in vitro by a direct cell-cell interaction. In vivo assay demonstrated that HECEC could significantly enhance the invasion and lung metastasis of esophageal cancer cells. To elucidate the molecular mechanisms of HECEC in esophageal carcinoma progression, we employed the microarray to analyze the gene expression profiles before and after treating with HECEC, HENEC or conditioned meium from HECEC. Among the highly expressed HECEC-regulated genes, we focused on Epiregulin (EREG). Further studies demonstrated that overexpression of EREG in EC9706 or Kyse30 cells can induce actin reorganization, sphere formation ability and a significantly enrichment of CD44+ cancer stem-like cells. Moreover, up-regulation of EREG in esophageal cancer cells could enhance lung metastasis and decrease the survival time in vivo. Further study indicated that EREG could induce activation of the Src and FAK. In addition, all these effects could also be inhibited by the function-blocking anti-EREG antibody in a dose dependent manner. Immunohistochemical analysis revealed that high level of EREG was significantly correlated with lymph node metastases and poor prognosis. In summary, HECEC play key roles in enhancing the invasion, migration, cancer stem cell phenotype and metastatic potential of esophageal cancer cells through Epiregulin.

12.
Metabolism ; 65(12): 1755-1767, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27832863

RESUMO

OBJECTIVE: Recent evidence has suggested that circulating endothelial progenitor cells (EPCs) can repair the arterial endothelium during vascular injury. However, a reliable source of human EPCs is needed for therapeutic applications. In this study, we isolated human fetal aorta (HFA)-derived EPCs and analyzed the capacity of EPCs to differentiate into endothelial cells. In addition, because microvascular dysfunction is considered to be the major cause of diabetic foot (DF), we investigated whether transplantation of HFA-derived EPCs could treat DF in a rat model. METHODS: EPCs were isolated from clinically aborted fetal aorta. RT-PCR, fluorescence-activated cell sorting, immunofluorescence, and an enzyme-linked immunosorbent assay were used to examine the expressions of CD133, CD34, CD31, Vascular Endothelial Growth Factor Receptor 2 (VEGFR2), von Willebrand Factor (vWF), and Endothelial Leukocyte Adhesion Molecule-1 (ELAM-1). Morphology and Dil-uptake were used to assess function of the EPCs. We then established a DF model by injecting microcarriers into the hind-limb arteries of Goto-Kakizaki rats and then transplanting the cultured EPCs into the ischemic hind limbs. Thermal infrared imaging, oxygen saturation apparatus, and laser Doppler perfusion imaging were used to monitor the progression of the disease. Immunohistochemistry was performed to examine the microvascular tissue formed by HFA-derived EPCs. RESULTS: We found that CD133, CD34, and VEGFR2 were expressed by HFA-derived EPCs. After VEGF induction, CD133 expression was significantly decreased, but expression levels of vWF and ELAM-1 were markedly increased. Furthermore, tube formation and Dil-uptake were improved after VEGF induction. These observations suggest that EPCs could differentiate into endothelial cells. In the DF model, temperature, blood flow, and oxygen saturation were reduced but recovered to a nearly normal level following injection of the EPCs in the hind limb. Ischemic symptoms also improved. Injected EPCs were preferentially and durably engrafted into the blood vessels. In addition, anti-human CD31+-AMA+-vWF+ microvasculars were detected after transplantation of EPCs. CONCLUSION: Early fetal aorta-derived EPCs possess strong self-renewal ability and can differentiate into endothelial cells. We demonstrated for the first time that transplanting HFA-derived EPCs could ameliorate DF prognosis in a rat model. These findings suggest that the transplantation of HFA-derived EPCs could serve as an innovative therapeutic strategy for managing DF.


Assuntos
Aorta/citologia , Transplante de Células/métodos , Pé Diabético/terapia , Células Progenitoras Endoteliais/citologia , Feto Abortado/citologia , Animais , Diferenciação Celular , Autorrenovação Celular , Células Endoteliais , Células Progenitoras Endoteliais/fisiologia , Células Progenitoras Endoteliais/transplante , Humanos , Masculino , Neovascularização Fisiológica , Ratos , Ratos Endogâmicos
13.
J Diabetes Res ; 2016: 9073037, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27635403

RESUMO

Advanced glycation end products (AGEs) are believed to be involved in diverse complications of diabetes mellitus. Overexposure to AGEs of pancreatic ß-cells leads to decreased insulin secretion and cell apoptosis. Here, to understand the cytotoxicity of AGEs to pancreatic ß-cells, we used INS-1-3 cells as a ß-cell model to address this question, which was a subclone of INS-1 cells and exhibited high level of insulin expression and high sensitivity to glucose stimulation. Exposed to large dose of AGEs, even though more insulin was synthesized, its secretion was significantly reduced from INS-1-3 cells. Further, AGEs treatment led to a time-dependent increase of depolymerized microtubules, which was accompanied by an increase of activated p38/MAPK in INS-1-3 cells. Pharmacological inhibition of p38/MAPK by SB202190 reversed microtubule depolymerization to a stabilized polymerization status but could not rescue the reduction of insulin release caused by AGEs. Taken together, these results suggest a novel role of AGEs-induced impairment of insulin secretion, which is partially due to a disturbance of microtubule dynamics that resulted from an activation of the p38/MAPK pathway.


Assuntos
Citoesqueleto/metabolismo , Glucose/farmacologia , Produtos Finais de Glicação Avançada/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Microtúbulos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Citoesqueleto/efeitos dos fármacos , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos
14.
Sci China Life Sci ; 59(3): 299-311, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26783141

RESUMO

Leukocyte transendothelial migration and sequestration are two distinct outcomes following leukocyte adhesion to endothelium during ischemia-reperfusion injury, in which platelets may play a pivotal role. In the present study, we established an in vitro hypoxia-reoxygenation model to mimic ischemia-reperfusion injury and found platelet pre-incubation significantly increased leukocyte adhesion to endothelial cells after hyoxia-reoxygenation (over 67%). Blockade of endothelial-cell-expressed adhesion molecules inhibited leukocyte direct adhesion to endothelial cells, while platelet-mediated leukocyte adhesion was suppressed by blockade of platelet-expressed adhesion molecules. Further experiments revealed platelets acted as a bridge to mediate leukocyte adhesion, and platelet-mediated adhesion was the predominant pattern in the presence of platelets. However, platelet pre-incubation significantly suppressed leukocyte transendothelial migration after hypoxia-reoxygenation (over 31%), which could be aggravated by blockade of endothelial-cell-expressed adhesion molecules, but alleviated by blockade of platelet- expressed adhesion molecules. This would indicate that platelet-mediated adhesion disrupted leukocyte transendothelial migration. An in vivo mesenteric ischemia-reperfusion model demonstrated leukocyte transfusion alone caused mild leukocyte adhesion to reperfused vessels and subsequent leukocyte infiltration, while simultaneous leukocyte and platelet transfusion led to massive leukocyte adhesion and sequestration within reperfused microvessels. Our studies revealed platelets enhanced leukocyte adhesion to endothelial cells, but suppressed leukocyte transendothelial migration. Overall, this leads to leukocyte sequestration in hypoxia-reoxygenated microvessels.


Assuntos
Plaquetas/metabolismo , Leucócitos/metabolismo , Microvasos/fisiopatologia , Traumatismo por Reperfusão/fisiopatologia , Plaquetas/citologia , Adesão Celular/fisiologia , Moléculas de Adesão Celular/metabolismo , Hipóxia Celular , Linhagem Celular , Movimento Celular , Células Cultivadas , Células Endoteliais/citologia , Humanos , Leucócitos/citologia , Microvasos/citologia , Modelos Biológicos
15.
Life Sci ; 144: 1-7, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26606859

RESUMO

AIMS: Glucocorticoids, such as dexamethasone, are widely used anti-inflammatory drugs. Their use is frequently associated with the development of steroid- associated diabetes. Pancreatic ß-cell dysfunction has been suggested to be one of the main causes of steroid-associated diabetes. However, the mechanism is not fully understood. Glycogen synthase kinase-3ß (GSK-3ß) is a multifunctional serine/threonine kinase and plays an important role in energy metabolism, cell growth and apoptosis. Therefore, the contribution of GSK-3ß in dexamethasone-induced pancreatic ß-cell apoptosis was determined in the present study. MAIN METHODS: The effect of dexamethasone treatment on rat pancreatic ß-cell line (INS-1) apoptosis (determined by TUNEL and Flow Cytometry), generation of reactive oxidative stress (ROS), and the phosphorylation status of GSK-3ß was determined. The inhibitory effect of GSK-3ß inhibitor-lithium chloride (LiCl) on dexamethasone-induced ß-cell apoptosis was also evaluated. KEY FINDINGS: Dexamethasone (0.1 µM) treatment induced INS-1 apoptosis, which was associated with increased GSK-3ß activation and increased NOX4-derived ROS generation. Pretreatment of INS-1 with LiCl inhibited dexamethasone induced ROS generation and INS-1 apoptosis. SIGNIFICANCE: This study provides a new mechanism of Dex induced pancreatic ß cell apoptosis and may serve as a new therapeutic option for treating GC induced diabetes.


Assuntos
Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Quinase 3 da Glicogênio Sintase/fisiologia , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Cloreto de Lítio/farmacologia , NADPH Oxidase 4 , NADPH Oxidases/biossíntese , NADPH Oxidases/genética , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Ratos , Espécies Reativas de Oxigênio/metabolismo
16.
Chin Med J (Engl) ; 128(24): 3357-62, 2015 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-26668152

RESUMO

BACKGROUND: Endothelial cell damage is an important pathophysiological step of restenosis after angioplasty and stenting. Cell transplantation has great therapeutic potential for endothelial recovery. We investigated the effect of transplanting endothelial progenitor cells (EPCs) derived from human early fetal aortas in rat injured arteries. METHODS: The carotid arterial endothelium of Sprague-Dawley rats was damaged by dilatation with a 1.5 F balloon catheter, and then EPCs derived from human early fetal aortas (<14 weeks) were injected into the lumen of the injured artery in transplanted rats, with an equal volume of normal saline injected into control rats. Rats were sacrificed at 2 and 4 weeks after treatment and transplanted cells were identified by immunohistochemical staining with anti-human CD31 and anti-human mitochondria antibodies. Arterial cross-sections were analyzed by pathology, immunohistochemistry, and morphometry. RESULTS: Green fluorescence-labeled EPCs could be seen in the endovascular surface of balloon-injured vessels after transplantation. The intimal area and intimal/medial area ratio were significantly smaller in the transplanted group than in the control (P < 0.05) and the residual lumen area was larger (P < 0.05). After EPC transplantation, a complete vascular endothelial layer was formed, which was positive for human von Willebrand factor after immunohistochemical staining, and immunohistochemical staining revealed many CD31- and mitochondria-positive cells in the re-endothelialized endothelium with EPC transplantation but not control treatment. CONCLUSION: EPCs derived from human early fetal aorta were successfully transplanted into injured vessels and might inhibit neointimal hyperplasia after vascular injury.


Assuntos
Artérias Carótidas/patologia , Transplante de Células , Células Progenitoras Endoteliais/citologia , Neointima/terapia , Animais , Adesão Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Progenitoras Endoteliais/fisiologia , Humanos , Imuno-Histoquímica , Microscopia de Fluorescência , Ratos , Ratos Sprague-Dawley
17.
Biochem Biophys Res Commun ; 461(4): 598-604, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-25912877

RESUMO

A significant portion of human and rat insulinomas coexpress multiple hormones. This character termed as multihormonality is also observed in some early pancreatic endocrine cells which coexpress insulin and glucagon, suggesting an incomplete differentiation status of both cells. Here we demonstrate that insulinoma cells INS-1 and INS-1-derived single cell clone INS-1-15 coexpressed insulin and glucagon in a portion of cells. These two hormones highly colocalized in the intracellular vesicles within a cell. Due to the existence of both PC1/3 and PC2 in INS-1-derived cells, proglucagon could be processed into glucagon, GLP-1 and GLP-2. These glucagon-family peptides and insulin were secreted simultaneously corresponding to the elevating glucose concentrations. The coexpression and partial colocalization of insulin and glucagon was also observed in rat fetal pancreatic endocrine cells, but the colocalization rate was generally lower and more diverse, suggesting that in the developing pancreatic endocrine cells, insulin and glucagon may be stored in nonidentical pools of secreting vesicles and might be secreted discordantly upon stimulus.


Assuntos
Células Endócrinas/metabolismo , Insulina/metabolismo , Insulinoma/metabolismo , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Frações Subcelulares/metabolismo , Animais , Linhagem Celular , Células Endócrinas/patologia , Glucagon , Insulinoma/patologia , Camundongos , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Ratos , Distribuição Tecidual
18.
Biochem Biophys Res Commun ; 458(3): 674-680, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25684188

RESUMO

Type 1 diabetes mellitus (T1DM) is characterized by the deficiencies of insulin and C-peptide. Mounting evidences have proved the beneficial effects of C-peptide on the renal function in T1DM. However, it is still controversial about the roles of C-peptide in T2DM nephropathy since the level of C-peptide fluctuates greatly at different stages of T2DM. In the present study, we found that the serum C-peptide concentration was much lower in GK rats with diabetic nephropathy than that in normal counterparts. A sustained supplementation of C-peptide at a physiological level could ameliorate urinary albumin, independent of blood glucose control. C-peptide treatment improved glomerulosclerosis and podocyte morphology and reduced the thickness of glomerular basement membrane as compared with saline treatment control. Moreover, it decreased fibronectin synthesis in diabetic glomeruli and in cultured rat mesangial cells accompanied by a down-regulation of RAGE and an up-regulation of PKA. Interestingly, H-89, a PKA inhibitor, could reverse the inhibition effect of C-peptide on fibronectin production in cultured mesangial cells. These findings suggest that C-peptide level is low in T2DM rats with nephropathy and a treatment with a physiological dose of C-peptide can prevent renal injury in diabetic GK rats.


Assuntos
Peptídeo C/uso terapêutico , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/tratamento farmacológico , Fibronectinas/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Albuminúria/complicações , Albuminúria/tratamento farmacológico , Animais , Peptídeo C/sangue , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Ativação Enzimática/efeitos dos fármacos , Fibronectinas/análise , Rim/metabolismo , Masculino , Ratos , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/análise , Receptores Imunológicos/metabolismo
19.
Biomaterials ; 37: 345-52, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25453963

RESUMO

Successful glioma gene therapy lays on two important factors, the therapeutic genes and efficient delivery vehicles to cross the blood-brain barrier (BBB) and reach gliomas. In this work, a new gene vector was constructed based on dendrigraft poly-l-lysines (DGL) and polyethyleneglycol (PEG), conjugated with a cell-penetrating peptide, the nucleolar translocation signal (NoLS) sequence of the LIM Kinase 2 (LIMK2) protein (LIMK2 NoLS peptide, LNP), yielding DGL-PEG-LNP. Plasmid DNA encoding inhibitor of growth 4 (ING4) was applied as the therapeutic gene. DGL-PEG-LNP/DNA nanoparticles (NPs) were monodispersed, with a mean diameter of 90.6 ± 8.9 nm. The conjugation of LNP significantly enhanced the BBB-crossing efficiency, cellular uptake and gene expression within tumor cells. Mechanism studies suggested the involvement of energy, caveolae-mediated endocytosis and macropinocytosis in cellular uptake of LNP-modified NPs. MTT results showed that no apparent cytotoxicity was observed when cells were treated with synthesized vectors. Furthermore, LNP-modified NPs mediated strongest and most intensive apoptosis on the tumor site, and the longest median survival time of glioma-bearing mice. All the results demonstrated that LNP is a kind of efficient CPPs especially for BBB-crossing application, and DGL-PEG-LNP/DNA is a potential non-viral platform for glioma gene therapy via intravenous administration.


Assuntos
Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Técnicas de Transferência de Genes , Glioma/tratamento farmacológico , Peptídeos/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Benzoxazóis/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Expressão Gênica , Glioma/patologia , Humanos , Masculino , Camundongos Nus , Nanopartículas/química , Peptídeos/farmacologia , Compostos de Quinolínio/metabolismo , Análise de Sobrevida
20.
Adv Healthc Mater ; 4(2): 291-300, 2015 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-25124929

RESUMO

Due to complication factors such as blood-brain barrier (BBB), integrating high efficiency of brain target ability with specific cargo releasing into one nanocarrier seems more important. A brain targeting nanoscale system is developed using dehydroascorbic acid (DHA) as targeting moiety. DHA has high affinity with GLUT1 on BBB. More importantly, the GLUT1 transportation of DHA represents a "one-way" accumulative priority from blood into brain. The artificial micelles are fabricated by a disulfide linkage, forming a bio-responsive inner barrier, which can maintain micelles highly stable in circulation and shield the leakage of entrapped drug before reaching the targeting cells. The designed micelles can cross BBB and be further internalized by brain cells. Once within the cells, the drug release can be triggered by high intracellular level of glutathione (GSH). Itraconazole (ITZ) is selected as the model drug because of its poor brain permeability and low stability in blood. It demonstrates that the functionalized nanoscale micelles can achieve highly effective direct drug delivery to targeting site. Based on the markedly increased stability in blood circulation and improved brain delivery efficiency of ITZ, DHA-modified micelles show highly effective in anti-intracranial infection. Therefore, this smart nanodevice shows a promising application for the treatment of brain diseases.


Assuntos
Anti-Infecciosos/farmacologia , Encéfalo/metabolismo , Encéfalo/patologia , Microambiente Celular , Micelas , Nanopartículas/química , Animais , Transporte Biológico/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Linhagem Celular Tumoral , Ácido Desidroascórbico/química , Ácido Desidroascórbico/metabolismo , Sistemas de Liberação de Medicamentos , Endocitose/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fluorescência , Glucose/metabolismo , Glutationa/metabolismo , Humanos , Terapia de Imunossupressão , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Microscopia de Força Atômica , Distribuição Tecidual/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...