Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Exp Mol Pathol ; 104(3): 250-255, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29621477

RESUMO

Diabetic wounds have impaired healing and a propensity for further morbidity, which may result in amputations. Stromal vascular fraction (SVF) is an autologous source of heterogeneous cell population obtained from adipose tissue, which is rich in stem cells and presents little immunogenicity to the host. In this study, we hypothesized that murine fibroblasts subjected to hyperglycemic conditions co-treated with SVF exhibit greater functional activity through the colorimetric MTT assay and a cell-monolayer in-vitro scratch assay. We sought to establish the underlying mechanism of action via the utility of an ELISA chemiluminescence array on the supernatant medium of the cells. Our results demonstrate that the mean percentage gap closure at 24 h in the hyperglycemia + SVF group was significantly greater at 41.1% ±â€¯1.6% compared to the hyperglycemia alone group 16.6% ±â€¯1.5% (post-hoc Bonferroni test p < 0.001, n = 3) although there was no difference between the SVF and normoglycemia group. Further, this SVF group exhibited a significantly greater 2.4 fold increase in fibroblastic cell viability as compared to the hyperglycemia alone group (p = 0.001, n = 3). The supernatant medium of the cells upon testing with ELISA indicated that early phase wound healing cytokines including platelet-derived growth factor (p = 0.012, n = 3), interleukin-1 (p = 0.003, n = 3), basic fibroblast growth factor (p = 0.003, n = 3) and interleukin-10 (p = 0.009, n = 3) were expressed in significantly greater relative luminescent units in SVF as compared to hyperglycemia alone groups (Student t-test). Taken together and for the first time, our study shows that SVF is a promising therapeutic agent for up-regulating fibroblastic activity in a hyperglycemic microenvironment, and this result can be explained in part by the stimulation of wound-healing cytokines.


Assuntos
Tecido Adiposo/patologia , Movimento Celular , Proliferação de Células , Citocinas/metabolismo , Fibroblastos/patologia , Hiperglicemia/patologia , Células Estromais/patologia , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/metabolismo , Hiperglicemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Estromais/metabolismo , Regulação para Cima , Cicatrização
2.
BMC Neurosci ; 13: 64, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22697290

RESUMO

BACKGROUND: Microglia, the resident immune cells of the central nervous system (CNS), have two distinct phenotypes in the developing brain: amoeboid form, known to be amoeboid microglial cells (AMC) and ramified form, known to be ramified microglial cells (RMC). The AMC are characterized by being proliferative, phagocytic and migratory whereas the RMC are quiescent and exhibit a slow turnover rate. The AMC transform into RMC with advancing age, and this transformation is indicative of the gradual shift in the microglial functions. Both AMC and RMC respond to CNS inflammation, and they become hypertrophic when activated by trauma, infection or neurodegenerative stimuli. The molecular mechanisms and functional significance of morphological transformation of microglia during normal development and in disease conditions is not clear. It is hypothesized that AMC and RMC are functionally regulated by a specific set of genes encoding various signaling molecules and transcription factors. RESULTS: To address this, we carried out cDNA microarray analysis using lectin-labeled AMC and RMC isolated from frozen tissue sections of the corpus callosum of 5-day and 4-week old rat brain respectively, by laser capture microdissection. The global gene expression profiles of both microglial phenotypes were compared and the differentially expressed genes in AMC and RMC were clustered based on their functional annotations. This genome wide comparative analysis identified genes that are specific to AMC and RMC. CONCLUSIONS: The novel and specific molecules identified from the trancriptome explains the quiescent state functioning of microglia in its two distinct morphological states.


Assuntos
Corpo Caloso/citologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Microglia/classificação , Microglia/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Linhagem Celular Transformada , Proliferação de Células , Análise por Conglomerados , Citocinas/genética , Citocinas/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Perfilação da Expressão Gênica , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Microdissecção e Captura a Laser , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Células-Tronco , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Histol Histopathol ; 26(8): 965-78, 2011 08.
Artigo em Inglês | MEDLINE | ID: mdl-21692030

RESUMO

BACKGROUND AND RESULTS: Embryos from diabetic mice exhibit several forms of neural tube defects, including non-closure of the neural tube. In the present study, embryos collected at embryonic day 11.5 from diabetic pregnancies displayed open neural tube with architectural disruption of the surrounding tissues. The percentage of proliferating cells was found to be increased in the dorsal and ventral domains of the spinal neural tube of embryos from diabetic mice, indicating a defect in the proliferation index. We have analyzed the development of various cell types, including motoneurons, interneurons, oligodendrocytes and migrating neurons, as well as radial glial cells in the open neural tube using specific molecular markers. Immunofluorescence results revealed a significantly reduced number of Pax2+ interneurons and increased number of Isl-1+ motoneurons, as well as Olig2+ oligodendrocytes in the neural tube of embryos from diabetic mice as compared to controls. In addition, these embryos exhibited a decreased number of doublecortin positive migrating neurons and Glast/Blbp positive radial glial cells with shortened processes in the neural tube. Expression levels of several developmental control genes involved in the generation of different neuronal cell types (such as Shh, Ngn, Ngn2, Ascl1) were also found to be altered in the neural tube of embryos from diabetic mice. CONCLUSIONS: Overall, the open neural tube in embryos of diabetic mice exhibits defects in the specification of different cell types, including motoneurons and interneurons, as well as glial cells along the dorsoventral axis of the developing spinal cord. Although these defects are associated with altered expression of several development control genes, the exact mechanisms by which maternal diabetes contributes to these changes remain to be investigated.


Assuntos
Diabetes Mellitus Experimental/patologia , Embrião de Mamíferos/anormalidades , Regulação da Expressão Gênica no Desenvolvimento , Defeitos do Tubo Neural/patologia , Tubo Neural/anormalidades , Gravidez em Diabéticas/patologia , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Idade Gestacional , Interneurônios/metabolismo , Interneurônios/patologia , Camundongos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Gravidez , Gravidez em Diabéticas/genética , Gravidez em Diabéticas/metabolismo
4.
Curr Med Chem ; 16(18): 2345-54, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19519395

RESUMO

Maternal diabetes develops in 2-6% of total pregnancies, depending on geographical and ethnic background. About 10% of fetuses from diabetic pregnancy display congenital malformations in various organ systems including cardiovascular, gastrointestinal, genitourinary and neurological systems, among which the neural tube defects (NTDs) such as anencephaly, holoprosencephaly and syntelencephaly were more frequently demonstrated. Recent studies by the Diabetes Control and Complications Trial Research Group show that tight glycemic control early in pregnancy decreases the progression of a number of diabetic complications. However, it appears that the pre-existing tissue damage cannot be reversed even after normoglycemic levels are achieved during pregnancy. In recent years, considerable efforts have been made to investigate the etiology of birth defects among infants of diabetic mothers. It has been shown that diabetes-induced fetal abnormalities are accompanied by some metabolic disturbances including elevated superoxide dismutase (SOD) activity, reduced levels of myoinositol and arachidonic acid and inhibition of the pentose phosphate shunt pathway. Moreover, the frequency of fetal malformations in diabetic pregnancy has been reported to be markedly reduced by dietary supplements of antioxidants such as vitamin E, vitamin C and butylated hy- droxytoluene, suggesting that oxidative stress is involved in the etiology of fetal dysmorphogenesis. Furthermore, several experimental studies have shown that NTDs in embryos of diabetic mice are associated with altered expression of genes, which control development of the neural tube. In this review, recent findings of possible molecular mechanisms which cause morphological changes during neural tube development in embryos of diabetic pregnancy are discussed.


Assuntos
Diabetes Gestacional/metabolismo , Defeitos do Tubo Neural/etiologia , Gravidez em Diabéticas/metabolismo , Animais , Apoptose , Encéfalo/embriologia , Encéfalo/metabolismo , Encéfalo/patologia , Diabetes Gestacional/epidemiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Tubo Neural/embriologia , Tubo Neural/metabolismo , Tubo Neural/patologia , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/genética , Estresse Oxidativo , Gravidez , Gravidez em Diabéticas/epidemiologia
5.
J Neurosci Res ; 86(16): 3481-93, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18655203

RESUMO

Maternal diabetes causes congenital malformations in various organs including the neural tube in fetuses. In this study, we have analyzed the differential gene expression profiling in the cranial neural tube of embryos from diabetic and control mice by using the oligonucleotide microarray. Expression patterns of genes and proteins that are differentially expressed in the cranial neural tube were further examined by the real-time reverse transcriptase-polymerase chain reaction, in situ hybridization, and immunohistochemistry. Proliferation index and apoptosis were examined by BrdU (5-bromo-2-deoxyuridine) labeling and TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling) assay, respectively. Embryos (E11.5) of diabetic pregnancies displayed distortion in neuroepithelia of the cranial neural tube. Microarray analysis revealed that a total of 390 genes exhibited more than twofold changes in expression level in the cranial neural tube of embryos from diabetic mice. Several genes involving apoptosis, proliferation, migration, and differentiation of neurons in the cranial neural tube were differentially expressed in embryos of diabetic pregnancy. In addition, maternal diabetes perturbed the development of choroid plexus and ventricular systems and reduced the production of proteins such as Ttr and Igf2 in the developing brain, indicating that these changes could impair the survival and proliferation of neuroepithelial cells and neurogenesis in embryos of diabetic mice. It is concluded that altered expression of a variety of genes involved in brain development is associated with cranial neural tube dysmorphogenesis that may subsequently contribute to intellectual impairment of the offspring of a diabetic mother.


Assuntos
Encéfalo/anormalidades , Complicações do Diabetes/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Defeitos do Tubo Neural/genética , Tubo Neural/anormalidades , Animais , Apoptose/genética , Padronização Corporal/genética , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Diferenciação Celular/genética , Linhagem da Célula/genética , Proliferação de Células , Ventrículos Cerebrais/anormalidades , Ventrículos Cerebrais/patologia , Ventrículos Cerebrais/fisiopatologia , Complicações do Diabetes/metabolismo , Complicações do Diabetes/fisiopatologia , Diabetes Mellitus Experimental/complicações , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/metabolismo , Tubo Neural/fisiopatologia , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/fisiopatologia , Neurônios/metabolismo , Gravidez , Células-Tronco/metabolismo
6.
Glia ; 50(1): 21-31, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15602748

RESUMO

The release of proinflammatory mediators such as tumor necrosis factor-alpha (TNF-alpha) and nitric oxide by microglia has been implicated in neurotoxicity in chronic neurodegenerative diseases such as Alzheimer's disease. As all-trans-retinoic acid (RA) has been reported to exert anti-inflammatory actions in various cell types, we have examined its effects on the expression of TNF-alpha and inducible nitric oxide synthase (iNOS) in microglia activated by beta-amyloid peptide (Abeta) and lipopolysaccharide (LPS). Exposure of primary cultures of rat microglial cells to Abeta or LPS stimulated the mRNA expression level of TNF-alpha (6-116-fold) and iNOS (8-500-fold) significantly. RA acted in a dose-dependent manner (0.1-10 microM) by attenuating both TNF-alpha (29-97%) and iNOS (61-96%) mRNA expression in microglia exposed to Abeta or LPS. RA-induced inhibition of TNF-alpha and iNOS mRNA expression in activated microglia was accompanied by the concomitant reduction in release of iNOS and TNF-alpha proteins as revealed by nitrite assay and ELISA, respectively. The anti-inflammatory effects of RA were correlated with the enhanced expression of retinoic acid receptor-beta, and transforming growth factor-beta1 as well as the inhibition of NF-kappaB translocation. These results suggest that RA may inhibit the neurotoxic effect of activated microglia by suppressing the production of inflammatory cytokines and cytotoxic molecules.


Assuntos
Microglia/metabolismo , Óxido Nítrico Sintase/biossíntese , Tretinoína/farmacologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Mediadores da Inflamação/farmacologia , Microglia/efeitos dos fármacos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Nitritos/análise , Nitritos/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do Ácido Retinoico/efeitos dos fármacos , Receptores do Ácido Retinoico/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética
7.
Clin Orthop Relat Res ; (411): 325-33, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12782891

RESUMO

Free muscle transfers do not generate the same force after transfer as that at the original sites. Light and electron microscopy were used to study serially during 30 weeks the changes at the neuromuscular junction after free muscle transfer of the gracilis muscle in the adult Wistar rat. Under light microscopy, after staining with acetylthiocholine the neuromuscular junction showed changes of degeneration with withdrawal of the innervating axon terminal followed by regeneration and reconstitution of the neuromuscular junction. The newly formed neuromuscular junction still lacked the structural detail seen in the control neuromuscular junction, even after 30 weeks. With the electron microscope, mitochondrial swelling and clumping of the synaptic vesicles were followed by withdrawal of the axon terminal from the muscle membrane on denervation. The infolding of the muscle membrane at the neuromuscular junction became less prominent. With reinnervation the ultrastructure of the junction was only partially reestablished with poorly reconstituted primary and secondary folds of the muscle membrane 30 weeks after the transfer. Failure of complete reformation of the ultrastructure of the neuromuscular junction may provide another explanation for failure of full recovery of skeletal muscle function after free muscle transfer.


Assuntos
Músculo Esquelético/transplante , Junção Neuromuscular/ultraestrutura , Animais , Feminino , Microscopia Eletrônica , Músculo Esquelético/inervação , Regeneração Nervosa , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA