Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Hum Genet ; 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38917801

RESUMEN

Extra-axial cavernous hemangiomas (ECHs) are complex vascular lesions mainly found in the spine and cavernous sinus. Their removal poses significant risk due to their vascularity and diffuse nature, and their genetic underpinnings remain incompletely understood. Our approach involved genetic analyses on 31 tissue samples of ECHs employing whole-exome sequencing and targeted deep sequencing. We explored downstream signaling pathways, gene expression changes, and resultant phenotypic shifts induced by these mutations, both in vitro and in vivo. In our cohort, 77.4% of samples had somatic missense variants in GNA14, GNAQ, or GJA4. Transcriptomic analysis highlighted significant pathway upregulation, with the GNAQ c.626A>G (p.Gln209Arg) mutation elevating PI3K-AKT-mTOR and angiogenesis-related pathways, while GNA14 c.614A>T (p.Gln205Leu) mutation led to MAPK and angiogenesis-related pathway upregulation. Using a mouse xenograft model, we observed enlarged vessels from these mutations. Additionally, we initiated rapamycin treatment in a 14-year-old individual harboring the GNAQ c.626A>G (p.Gln209Arg) variant, resulting in gradual regression of cutaneous cavernous hemangiomas and improved motor strength, with minimal side effects. Understanding these mutations and their pathways provides a foundation for developing therapies for ECHs resistant to current therapies. Indeed, the administration of rapamycin in an individual within this study highlights the promise of targeted treatments in treating these complex lesions.

2.
Cell Physiol Biochem ; 46(5): 1951-1970, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29719282

RESUMEN

BACKGROUND/AIMS: Neurotrophic effects and immunosuppression are the main therapeutic mechanisms of mesenchymal stem cells (MSCs) in stroke treatment. Neurotrophins are produced by graft cells, host neurons, astrocytes, and even microglia/macrophages. Meanwhile, MSCs can increase inflammation if they are not sufficiently induced by pro-inflammatory cytokines. We examined whether intravenously transplanted bone marrow MSCs (BM-MSCs) increase inflammation in distal middle cerebral artery occlusion (dMCAO) rats, how long the increased inflammation effect persists for, and what the final therapeutic outcomes will be. We also tested the neurotrophic role of BM-MSCs and attempted to identify the neurotrophin-producing cells. METHODS: At 1 h after dMCAO was performed on Sprague-Dawley rats, allogeneic BM-MSCs were transplanted intravenously. The infarct volume was examined by Tetrazolium Red staining at 2 days (day 2), and the behavioral tests (cylinder test and grid walking test) were performed at 2, 4 (day 4) and 7 days (day 7) after transplantation. The concentrations of inflammation related cytokines and neurotrophins in the ischemic cortex, ipsilateral striatum, and serum, were measured using ELISA at days 2-7. The cell source of neurotrophins was observed by immunohistochemistry. RESULTS: The transplanted cells were mainly found in the infarct border zone (IBZ) of the brain. Infarct volume was reduced and behavioral outcomes were improved at 2 days after ischemia. In the striatum and circulation, BM-MSC transplantation increased inflammation at day 2 and decreased it at day 7. At days 2-7, insulin-like growth factor-1 (IGF-1) and brain-derived neurotrophic factor (BDNF) concentrations in the ischemic core of the cortex were significantly higher in the BM-MSC group than in the ischemia vehicle group. IGF-1 and BDNF were derived mainly from host microglia/macrophages in the ischemic core, and transplanted cells in the IBZ. At day 2, BM-MSC transplantation significantly increased the number of IGF-1+CD68+ and BDNF+Iba-1+ double positive cells in the ischemic core cortex. CONCLUSIONS: Although increased inflammation, BM-MSCs were still beneficial to dMCAO recovery at day 2. The immunopromoting effect of MSCs was transient and shifted to an immunosuppressive action at day 7. The neurotrophic factors IGF-1 and BDNF, which were mainly derived from transplanted BM-MSCs and host microglia/macrophages, contributed to the therapeutic effects from day 2 to day 7.


Asunto(s)
Infarto de la Arteria Cerebral Media/terapia , Inflamación/etiología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Trasplante de Células Madre Mesenquimatosas/métodos , Administración Intravenosa , Animales , Movimiento Celular , Células Cultivadas , Citocinas/análisis , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Masculino , Ratas Sprague-Dawley
3.
Cell Physiol Biochem ; 41(2): 819-834, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28214829

RESUMEN

BACKGROUND/AIMS: EphB4 belongs to the largest family of Eph receptor tyrosine kinases. It contributes to a variety of pathological progresses of cancer malignancy. However, little is known about its role in neural stem cells (NSCs). This study examined whether EphB4 is required for proliferation and differentiation of human embryonic neural stem cells (hNSCs) in vitro. METHODS: We up- and down-regulated EphB4 expression in hNSCs using lentiviral over-expression and shRNA knockdown constructs and then investigated the influence of EphB4 on the properties of hNSCs. RESULTS: Our results show that shRNA-mediated EphB4 reduction profoundly impaired hNSCs self-renewal and proliferation. Furthermore, detection of differentiation revealed that knockdown of EphB4 inhibited hNSCs differentiation towards a neuronal lineage and promoted hNSCs differentiation to glial cells. In contrast, EphB4 overexpression promoted hNSCs self-renewal and proliferation, further induced hNSCs differentiation towards a neuronal lineage and inhibited hNSCs differentiation to glial cells. Moreover, we found that EphB4 regulates cell proliferation mediated by the Abl-CyclinD1 pathway. CONCLUSION: These studies provide strong evidence that fine tuning of EphB4 expression is crucial for the proliferation and neuronal differentiation of hNSCs, suggesting that EphB4 might be an interesting target for overcoming some of the therapeutic limitations of neuronal loss in brain diseases.


Asunto(s)
Células-Madre Neurales/metabolismo , Receptor EphB4/metabolismo , Puntos de Control del Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Humanos , Células-Madre Neurales/citología , Neuronas/citología , Proteínas Proto-Oncogénicas c-abl/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor EphB4/antagonistas & inhibidores , Receptor EphB4/genética , Transducción de Señal
4.
Cell Physiol Biochem ; 38(6): 2123-38, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27309670

RESUMEN

BACKGROUND: Human fetal striatum-derived neural stem cells (hfsNSCs) are important in regenerative medicine; however, their ability to self-renew diminishes quickly following passages in culture. Typically when hfsNSC-derived neurospheres are dissociated by accutase, more than 90% of the cells survive, but only 6-8% of the cells are able to form secondary neurospheres. Our hypothesis is that the hfsNSCs that are unable to form new neurospheres become apoptotic. METHODS/RESULTS: Because the NSC apoptosis process has never been characterized in detail, we characterized hfsNSC apoptosis using multiparameter analysis and determined that the majority of hfsNSCs undergo apoptosis after passaging, which leads to a reduction in self-renewal. The replacement of trituration with vortexing decreases apoptosis, increases self-renewal, and does not affect NSC differentiation. When we used live cell staining with Annexin V, Hoechst 33342, and PI together, the apoptotic index was in agreement with what could be obtained using fixed-cell staining methods, including TUNEL and activated caspase-3 immunocytochemistry. NSC apoptosis could be divided into 9 stage types based on our live cell assay. Several types during early and late stages had similar staining profiles that could be further discriminated based on cell size. CONCLUSION: Apoptosis largely contributes to the low self-renewal of neurospheres, and replacing trituration with vortexing aided in alleviating NSC apoptosis. Multiparameter analysis is required for the identification of NSC apoptosis, particularly when live cell staining is used.


Asunto(s)
Apoptosis , Feto/citología , Células-Madre Neurales/citología , Caspasa 3/análisis , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos , Etiquetado Corte-Fin in Situ
5.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 38(5): 497-506, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27825404

RESUMEN

Objective To compare the effecacy of human mesenchymal stromal cell (hMSC) with human mononuclear cell (hMNC) in treating rat cerebral infarct.Methods The SD rat models of cerebral infarct were established by distal middle cerebral artery occlusion (dMCAO). Rats were divided into four groups: sham,ischemia vehicle,MSC,and MNC transplantation groups. For the transplantation group,1×106 hMSCs or hMNCs were intravascularly transplanted into the tail vein 1 hour after the ischemia onset. The ischemia vehicle group received dMCAO surgery and intravascular saline injection 1,3,5,and 7 days after the ischemia onset,and then behavioral tests were performed. At 48 h after the ischemia onset,the abundance of Iba- 1,the symbol of activated microglia,was evaluated in the peri-ischemia striatum area; meanwhile,the neurotrophic factors such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) in ipsilateral peri-ischemia striatum area were also measured. Results The relative infarct volume in ischemia vehicle group,hMSC group,and hMNC transplantation group were (37.85±4.40)%,(33.41±3.82)%,and (30.23±3.63)%,respectively. The infarct volumes of MSC group (t=2.100,P=0.034) and MNC group (t=2.109,P=0.0009) were significantly smaller than that of ischemia vehicle group,and that of MNC group was significantly smaller than that of MSC group (t=1.743,P=0.043). One day after transplantation,the score of ischemia vehicle group in limb placing test was (4.32±0.71)%,which was significantly lower than that in sham group (9.73±0.36)% (t=2.178,P=8.61×10-11). The scores of MSC and MNC group,which were (5.09±0.62)% (t=2.1009,P=0.024) and (5.90±0.68)% (t=2.1008,P=0.0001),respectively,were significantly higher than that of ischemia vehicle group; also,the score of MNC group was significantly higher than that of MSC group(t=2.1009,P=0.0165). The contralateral forelimb scores of MSC and MNC groups in beam walking test were (5.56±0.86)% (t=2.120,P=0.020) and (5.13±0.95)% (t=2.131,P=0.003),were both significantly lower than that of ischemia vehicle group [(6.47±0.61)%]. Three days after the transplantation,the limb placing test score of MNC group [(6.91±1.10)%] was significantly higher than that of ischemia vehicle group (5.80±0.82)% (t=2.110,P=0.027). The score of MSC group [(6.30±0.77)%] showed no statistic difference with that of ischemia vehicle group(t=2.101,P=0.199).The contralateral forelimb scores of MNC group in beam walking test [(4.34±0.58)%] was significantly lower than that of ischemia vehicle group [(5.31±0.65)%] (t=2.100,P=0.006) and MSC group [(4.92±0.53)%] (t=2.100,P=0.041); there was no statistic difference between MSC group and ischemia vehicle group (t=2.109,P=0.139). The relative abundance of Iba- 1 in sham,ischemia vehicle,MSC,and MNC groups was 1.00+0.00,1.72±0.21,1.23±0.08,and 1.48±0.06,respectively. The Iba-1 relative abundance of ischemia vehicle group was significantly higher than that of sham group (t=2.262,P=2.9×10-6). The Iba-1 relative abundances of both MSC (t=2.178,P=3.91×10-5)and MNC (t=2.200,P=0.007)groups were significantly lower than that of ischemia vehicle group. It was also significantly lower in MNC group than in MSC group also (t=2.120,P=7.09×10-6). Three days after transplantation,the BDNF and GDNF levels of MSC group,which were (531.127±73.176)pg/mg (t=2.109,P=0.003)and(127.780±16.733)pg/mg(t=2.100,P=2.76×10-5),respectively,were significantly higher than those of ischemia vehicle group,which were (401.988±89.006)pg/mg and (86.278±14.832) pg/mg,respectively. The BDNF and GDNF levels of MNC group,which were (627.429±65.646)pg/mg (t=2.144,P=0.017) and (153.117±20.443)pg/mg (t=2.109,P=0.010),respectively,were all significantly higher than that of MSC group. At day 7,the BDNF and GDNF levels of MSC group,which were (504.776±83.282)pg/mg (t=2.101,P=0.005) and (81.641±11.019)pg/mg (t=2.100,P=0.002),respectively,were significantly higher than those of ischemia vehicle group,which were (389.257±70.440)pg/mg and (64.322±9.855) pg/mg,respectively. The BDNF and GDNF levels of MNC group,which were (589.068±63.323)pg/mg (t=2.100,P=0.027) and (102.161±19.932)pg/mg (t=2.144,P=0.017),respectively,were all significantly higher than that of MSC group. Conclusions Both hMSC and hMNC are beneficial to the ischemia-damaged brain when they are intravascularly transplanted within 1 h after the onset of ischemia. The anti-inflammation ability and secretion of neurotrophic factors are the underlying mechanisms of the therapeutic effects. MNC is more effective than MSC in reducing infarct area and improving behaviors,which might be explained by the fact that MNC induces more GDNF and BDNF in brain than MSC.


Asunto(s)
Isquemia Encefálica/terapia , Infarto de la Arteria Cerebral Media/terapia , Leucocitos Mononucleares/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Médula Ósea , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Feto , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
6.
J Neurosci Res ; 92(1): 74-85, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24123213

RESUMEN

Neural progenitor cells (NPCs) derived from mouse embryonic stem (mES) cells can lead to tumors after transplantation. The cellular source of such tumors remains under debate. We investigated the tumor formation resulting from mES cell-derived NPCs in a rat stroke model and in nude mice. After 2 hr of ischemia and 48 hr of reperfusion, the NPCs were transplanted into the ischemic core of the xenogeneic rats. Four weeks after transplantation, the grafted cells were found to be viable at the border of the necrosis and had differentiated into neurons. Transplanted rats did not exhibit any behavioral improvement, because tumor formed in 90% of the animals. Immunosuppression facilitated tumor formation. Tumors were observed in 40% of normal rats after NPC transplantation when cyclosporin A was administered. Meanwhile, no tumor formation was observed without cyclosporin A. Ischemic damage also facilitated tumor formation, because NPCs gave rise to tumors in 90% of ischemic rats, a percentage significantly higher than that in intact rats, which was 40%. The SSEA-1-positive cells isolated from stage 4 are not exactly undifferentiated ES cells. They exhibited a marker gene transcription profile different from that of ES cells and did not form tumors in transplanted nude mice. The undifferentiated ES cells remaining after differentiation did not contribute to tumors either. First, the tumor formation rate resulting from undifferentiated ES cells in the brains of normal rats is 0%, significantly lower than that of NPCs. Second, transplanted NPCs that led to 100% tumors in nude mice contained approximately 1.5 × 10(3) Oct-4-positive cells; however, even 5 × 10(5) undifferentiated ES cells formed neoplasm only in 40% nude mice.


Asunto(s)
Isquemia Encefálica/patología , Neoplasias Encefálicas/patología , Células Madre Embrionarias/trasplante , Terapia de Inmunosupresión , Antígeno Lewis X/metabolismo , Células-Madre Neurales/trasplante , Animales , Isquemia Encefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Recuento de Células , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Masculino , Ratones , Ratones Desnudos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Ratas , Ratas Wistar , Trasplante de Células Madre/métodos
7.
Tissue Eng Regen Med ; 21(4): 625-639, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38578425

RESUMEN

BACKGROUND: Syringomyelia is a progressive chronic disease that leads to nerve pain, sensory dissociation, and dyskinesia. Symptoms often do not improve after surgery. Stem cells have been widely explored for the treatment of nervous system diseases due to their immunoregulatory and neural replacement abilities. METHODS: In this study, we used a rat model of syringomyelia characterized by focal dilatation of the central canal to explore an effective transplantation scheme and evaluate the effect of mesenchymal stem cells and induced neural stem cells for the treatment of syringomyelia. RESULTS: The results showed that cell transplantation could not only promote syrinx shrinkage but also stimulate the proliferation of ependymal cells, and the effect of this result was related to the transplantation location. These reactions appeared only when the cells were transplanted into the cavity. Additionally, we discovered that cell transplantation transformed activated microglia into the M2 phenotype. IGF1-expressing M2 microglia may play a significant role in the repair of nerve pain. CONCLUSION: Cell transplantation can promote cavity shrinkage and regulate the local inflammatory environment. Moreover, the proliferation of ependymal cells may indicate the activation of endogenous stem cells, which is important for the regeneration and repair of spinal cord injury.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Células-Madre Neurales , Ratas Sprague-Dawley , Siringomielia , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Siringomielia/terapia , Ratas , Proliferación Celular , Epéndimo , Masculino , Microglía/metabolismo , Modelos Animales de Enfermedad
8.
J Cell Biochem ; 114(7): 1549-58, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23297156

RESUMEN

Folate is thought to contribute to health and development by methylation regulation. Long interspersed nucleotide element-1 (LINE-1), which is regulated by methylation modification, plays an important role in sculpting the structure and function of genomes. Some studies have shown that folate concentration is related to LINE-1 methylation. However, the direct association between LINE-1 methylation and folate deficiency remains unclear. To explore whether folate deficiency directly induced LINE-1 hypomethylation and to analyze the relationship between folate concentration and the LINE-1 methylation level, mouse ESCs were treated with various concentrations of folate which was measured by chemiluminescent immunoassay, and the homocysteine content was detected by ELISA. LINE-1 methylation was examined by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry at various time points. Concurrently, cell proliferation and differentiation were observed. The result showed that the intracellular folate decreases under folate-deficient condition, conversely, homocysteine content increased gradually and there was a negatively correlated between them. Folate insufficiency induced LINE-1 hypomethylation at the lowest levels in folate-free group and moderate in folate-deficient group, compared with that in the folate-normal group at day 18. Moreover, LINE-1 methylation level was positively correlated with folate content, and negatively correlated with homocysteine content. At corresponding time points, proliferation and differentiation of mouse ESCs showed no alteration in all groups. Our data indicated that folate deficiency affected the homeostasis of folate-mediated one-carbon metabolism, leading to reduced LINE-1 methylation in mouse ESCs. This study provides preliminary evidence of folate deficiency affecting early embryonic development.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Ácido Fólico/farmacología , Elementos de Nucleótido Esparcido Largo/genética , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Metilación de ADN/efectos de los fármacos , Células Madre Embrionarias/citología , Ratones
9.
Zhonghua Yi Xue Za Zhi ; 93(13): 1012-5, 2013 Apr 02.
Artículo en Zh | MEDLINE | ID: mdl-23886267

RESUMEN

OBJECTIVE: To assess the sensitivity and specificity of anti-aquaporin 4 antibody in the diagnosis of neuromyelitis optical (NMO) and analyze the relationship between clinical features and different NMO-IgG status. METHODS: A total of 269 serum specimens were collected from the patients with NMO, high-risk for NMO, multiple sclerosis (MS) and miscellaneous diseases and analyzed with HEK-293T cells transfected by aquaporin 4 cDNA. The sensitivity and specificity of anti-aquaporin 4 antibody in the diagnosis of NMO were calculated, Spearman correlation coefficients were used to examine the relationship between clinical features and antibody titer. RESULTS: (1) The results of cell-based immunofluorescence assay showed 36 examples of 47 NMO patient serums (76.6%) were positive, 7/23 high-risk for NMO positive (30.4%), 3/85 multiple sclerosis (3.5%) positive, 1/48 miscellaneous neurological disorders (2.1%), 2/16 immune system diseases positive (12.5%) and negative in all 50 healthy serum specimens. Sensitivity and specificity were 76.6% and 97.0% for discriminating NMO from MS and other diseases. (2) Anti-AQP4 antibody titer had no obvious correlation to relapses, spinal lesion length and EDSS (P > 0.05). (3) Anti-AQP4 antibody titer became lower after a high dose of intravenous methylprednisolone (P < 0.05). CONCLUSION: Anti-AQP4 antibody in NMO has a high specificity and sensitivity so as to contribute to early diagnosis and optimized treatment of NMO. And its titer decreases after a high dose of intravenous methylprednisolone. But there is no correlation between its titer and length of spinal cord lesions, relapsing frequency or expanded disability status scale (EDSS) score.


Asunto(s)
Acuaporina 4/inmunología , Autoanticuerpos/inmunología , Neuromielitis Óptica/diagnóstico , Neuromielitis Óptica/inmunología , Adulto , Acuaporina 4/sangre , Autoanticuerpos/sangre , Femenino , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Neuromielitis Óptica/terapia , Sensibilidad y Especificidad
11.
Exp Cell Res ; 317(20): 2950-7, 2011 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21963525

RESUMEN

Mesenchymal stem cells (MSCs) have shown potential clinical utility in cell therapy and tissue engineering, due to their ability to proliferate as well as to differentiate into multiple lineages, including osteogenic, adipogenic, and chondrogenic specifications. Therefore, it is crucial to assess the safety of MSCs while extensive expansion ex vivo is a prerequisite to obtain the cell numbers for cell transplantation. Here we show that MSCs derived from adult cynomolgus monkey can undergo spontaneous transformation following in vitro culture. In comparison with MSCs, the spontaneously transformed mesenchymal cells (TMCs) display significantly different growth pattern and morphology, reminiscent of the characteristics of tumor cells. Importantly, TMCs are highly tumorigenic, causing subcutaneous tumors when injected into NOD/SCID mice. Moreover, no multiple differentiation potential of TMCs is observed in vitro or in vivo, suggesting that spontaneously transformed adult stem cells may not necessarily turn into cancer stem cells. These data indicate a direct transformation of cynomolgus monkey MSCs into tumor cells following long-term expansion in vitro. The spontaneous transformation of the cultured cynomolgus monkey MSCs may have important implications for ongoing clinical trials and for models of oncogenesis, thus warranting a more strict assessment of MSCs prior to cell therapy.


Asunto(s)
Células Madre Adultas/patología , Transformación Celular Neoplásica/patología , Células Madre Mesenquimatosas/patología , Animales , Antígenos de Superficie/genética , Diferenciación Celular/fisiología , Transformación Celular Neoplásica/genética , Cariotipo , Macaca , Macaca fascicularis , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Telomerasa/metabolismo
12.
Front Genet ; 13: 852049, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35591854

RESUMEN

Background: Glioma is globally recognised as one of the most frequently occurring primary malignant brain tumours, making the identification of glioma biomarkers critically significant. The protein KIF18A (Kinesin Family Member 18A) is a member of the kinesin superfamily of microtubule-associated molecular motors and has been shown to participate in cell cycle and mitotic metaphase and anaphase. This is the first investigation into the expression of KIF18A and its prognostic value, potential biological functions, and effects on the immune system and mitosis in glioma patients. Methods: Gene expression and clinicopathological analysis, enrichment analysis, and immune infiltration analysis were based on data obtained from The Cancer Genome Atlas (TCGA), with additional bioinformatics analyses performed. Statistical analysis was conducted in R software. Clinical samples were used to evaluate the expression of KIF18A via immunohistochemical staining. In addition, the expression level of KIF18A was validated on U87 cell line. Results: Our results highlighted that KIF18A plays a key role as an independent prognostic factor in patients with glioma. KIF18A was highly expressed in glioma tissues, and KIF18A expression was associated with age, World Health Organization grade, isocitrate dehydrogenase (IDH) status, 1p/19q codeletion, primary therapy outcome, and overall survival (OS). Enrichment analysis revealed that KIF18A is closely correlated with the cell cycle and mitosis. Single sample gene set enrichment analysis (ssGSEA) analysis revealed that KIF18A expression was related to the immune microenvironment. The increased expression of KIF18A in glioma was verified in clinical samples and U87 cell line. Conclusion: The identification of KIF18A as a new biomarker for glioma could help elucidate how changes in the glioma cell and immune microenvironment promote glioma malignancy. With further analysis, KIF18A may serve as an independent prognostic indicator for human glioma.

13.
Acta Histochem ; 124(6): 151927, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35792494

RESUMEN

Induced neural stem cells (iNSCs) can be reprogrammed from somatic cells and have shown potentials in treatment of various neurological diseases/disorders. Obtaining iNSCs of nonhuman primates serves as an important bridge for clinical translation using iNSCs. In the current study, cynomolgus (Macaca fascicularis) bone marrow mesenchymal stromal cells (MSCs) were reprogrammed into iNSCs by transduction of non-integrative Sendai virus encoding transgenes OCT4, SOX2, KLF4 and C-MYC. The obtained iNSCs showed characteristics of normal neural stem cells (NSCs) and could differentiate into neurons, astrocytes and oligodendrocytes. Furthermore, iNSCs could give rise to dopaminergic neural cells in vitro, which showed safety and efficacy after transplantation into the striatum of an immunodeficient mouse Parkinson's disease (PD) model.


Asunto(s)
Células-Madre Neurales , Enfermedad de Parkinson , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Macaca fascicularis , Ratones , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia
14.
Front Immunol ; 13: 845223, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35350788

RESUMEN

Glioma represents the most common primary intracranial malignancy worldwide, with low overall survival rates and limited therapeutic options. The protein CD101, mainly expressed on several immune cells, has been demonstrated to exert potent effects on blunting T cell immune responses across infectious and autoimmunity diseases. Nevertheless, the prognostic value of CD101 expression and its role in the immune microenvironment of various malignancies currently remains elusive. Herein, by adopting bioinformatics methodology, we comprehensively illustrated the potential function and predictive value of CD101 in stratifying clinical prognosis among patients with glioma, for which a high CD101 level predicted an unfavorable clinical outcome in glioma patients. Results from enrichment analyses manifested that CD101 predominantly expressed on the tumor-associated macrophages and was significantly associated with the immune regulatory processes, as evidenced by its positive correlation with immune-related genes and the putative infiltration of immune cells. Evidence provided by in-situ multicolor immunofluorescence staining further validated our findings at the protein level. Taken together, CD101 may serve as a novel biomarker in predicting clinical prognosis and immune status for glioma patients.


Asunto(s)
Neoplasias Encefálicas , Glioma , Biomarcadores , Neoplasias Encefálicas/patología , Glioma/patología , Humanos , Macrófagos , Pronóstico , Microambiente Tumoral
15.
Front Neurosci ; 16: 1035444, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36760798

RESUMEN

Introduction: Parkinson's disease (PD), as a common neurodegenerative disease, currently has no effective therapeutic approaches to delay or stop its progression. There is an urgent need to further define its pathogenesis and develop new therapeutic targets. An increasing number of studies have shown that members of the sirtuin (SIRT) family are differentially involved in neurodegenerative diseases, indicating their potential to serve as targets in therapeutic strategies. Mitochondrial SIRT4 possesses multiple enzymatic activities, such as deacetylase, ADP ribosyltransferase, lipoamidase, and deacylase activities, and exhibits different enzymatic activities and target substrates in different tissues and cells; thus, mitochondrial SIRT4 plays an integral role in regulating metabolism. However, the role and mechanism of SIRT4 in PD are not fully understood. This study aimed to investigate the potential mechanism and possible regulatory targets of SIRT4 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice. Methods: The expression of the SIRT4 protein in the MPTP-induced PD mouse mice or key familial Parkinson disease protein 7 knockout (DJ-1 KO) rat was compared against the control group by western blot assay. Afterwards, quantitative proteomics and bioinformatics analyses were performed to identify altered proteins in the vitro model and reveal the possible functional role of SIRT4. The most promising molecular target of SIRT4 were screened and validated by viral transfection, western blot assay and reverse transcription quantitative PCR (RT-qPCR) assays. Results: The expression of the SIRT4 protein was found to be altered both in the MPTP-induced PD mouse mice and DJ-1KO rats. Following the viral transfection of SIRT4, a quantitative proteomics analysis identified 5,094 altered proteins in the vitro model, including 213 significantly upregulated proteins and 222 significantly downregulated proteins. The results from bioinformatics analyses indicated that SIRT4 mainly affected the ribosomal pathway, propionate metabolism pathway, peroxisome proliferator-activated receptor (PPAR) signaling pathway and peroxisome pathway in cells, and we screened 25 potential molecular targets. Finally, only fatty acid binding protein 4 (FABP4) in the PPAR signaling pathway was regulated by SIRT4 among the 25 molecules. Importantly, the alterations in FABP4 and PPARγ were verified in the MPTP-induced PD mouse model. Discussion: Our results indicated that FABP4 in the PPAR signaling pathway is the most promising molecular target of SIRT4 in an MPTP-induced mouse model and revealed the possible functional role of SIRT4. This study provides a reference for future drug development and mechanism research with SIRT4 as a target or biomarker.

16.
Adv Mater ; 34(50): e2205881, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36189858

RESUMEN

Gas therapy is an emerging technology for improving cancer therapy with high efficiency and low side effects. However, due to the existence of the gatekeeper of the blood-brain barrier (BBB) and the limited availability of current drug delivery systems, there still have been no reports on gas therapy for intracranial neuroglioma. Herein, an integrated, self-powered, and wirelessly controlled gas-therapy system is reported, which is composed of a self-powered triboelectric nanogenerator (TENG) and an implantable nitric oxide (NO) releasing device for intracranial neuroglioma therapy. In the system, the patient self-driven TENG converts the mechanical energy of body movements into electricity as a sustainable and self-controlled power source. When delivering energy to light a light-emitting diode in the implantable NO releasing device via wireless control, the encapsulated NO donor s-nitrosoglutathione (GSNO) can generate NO gas to locally kill the glioma cells. The efficacy of the proof-of-concept system in subcutaneous 4T1 breast cancer model in mice and intracranial glioblastoma multiforme in rats is verified. This self-powered gas-therapy system has great potential to be an effective adjuvant treatment modality to inhibit tumor growth, relapse, and invasion via teletherapy.


Asunto(s)
Nanotecnología , Óxido Nítrico , Ratas , Ratones , Animales , Recurrencia Local de Neoplasia , Suministros de Energía Eléctrica , Electricidad
17.
Zhonghua Yi Xue Za Zhi ; 91(16): 1122-4, 2011 Apr 26.
Artículo en Zh | MEDLINE | ID: mdl-21609597

RESUMEN

OBJECTIVE: To ascertain the diagnosis of such a rare disease as Ehlers-Danlos syndrome type IV by the technique of DNA(deoxyribonucleic acid)analysis. METHODS: The primer sequences of Col3A1 gene were designed. Genomic DNA was isolated from the peripheral blood samples. The amplification of polymerase chain reaction (PCR) was performed and direct sequencing used to screen the mutations. A definite diagnosis was made in conjunctions with clinical features. RESULTS: Two nucleotide mutations for Col3A1 were found. One was in intron 15 while another in exon 30. The latter was an important mutation of a G to A transition (c.2209G > A) resulting in alanine to threonine substitution at position (p.Ala698Thr). The mutations were inherited from proband of pedigree. CONCLUSION: Genetic testing of Col3A1 mutation can facilitate an accurate diagnosis of Ehlers-Danlos syndrome.


Asunto(s)
Colágeno Tipo III/genética , Síndrome de Ehlers-Danlos/diagnóstico , Síndrome de Ehlers-Danlos/genética , Mutación , Niño , Femenino , Humanos
18.
Biomed Pharmacother ; 142: 111971, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34343893

RESUMEN

Recent studies report that inhibiting TNF-α might be a novel therapeutic strategy for managing brain ischemia. Our previous study reported that mesenchymal stem cell (MSC) transplantation could suppress TNF-α level in both serum and brain. However, the cell type(s) that contribute to the production of TNF-α during ischemia following MSC transplantation has not been well studied. In the present study, we found by fluorescent immunohistochemistry, that 7.95 ± 6.17% of TNF-α+ cells co-expressed Iba-1 in the infarct area of dMCAO rats, a majority of which were found to be CD68+ (activated microglia), suggesting that resident microglial population were not the major source of TNF-α expression. 68.49 ± 5.12% of the TNF-α+ cells in the infarct area could be labeled by GFAP, a specific marker for astrocytes, indicating that resident GFAP+ astrocytes might be the major source of TNF-α expression in the infarct area. In addition to the infarct area, the GFAP+/TNF-α+ double-positive astrocytes accounted for 73.68 ± 7.48% of the TNF-α+ cells in striatum and corpus callosum. The infiltrating cells, including monocytes and lymphocytes, were not the main source of TNF-α either. In response to MSC transplantation, the total TNF-α+ cells as well as the percentage of TNF-α-expressing astrocytes were significantly reduced in the infarct area, suggesting that MSC transplantation could suppress the expression of TNF-α by astrocytes. Taken together, the results demonstrated that resident astrocytes, but not microglia, were the major source of TNF-α expression and could be suppressed by MSC infusion.


Asunto(s)
Astrocitos/citología , Infarto Encefálico/fisiopatología , Trasplante de Células Madre Mesenquimatosas/métodos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Infarto Encefálico/terapia , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/terapia , Modelos Animales de Enfermedad , Inmunohistoquímica , Infarto de la Arteria Cerebral Media , Masculino , Microglía/citología , Ratas , Ratas Sprague-Dawley
19.
J Assist Reprod Genet ; 27(6): 285-91, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20393797

RESUMEN

PURPOSE: Feeder cells from animals raise considerable concern for contamination because they are directly in contact with embryonic stem cells. METHODS: To address this issue we collected discarded foreskin tissue and prepared a fibroblast cell line. We transferred one parthenogenetic blastocyst on to these feeder cells, and later observed outgrowth. By this approach, we were able to derive a human parthenogenetic embryonic stem cell line successfully. RESULTS: The embryonic stem cells had normal morphology, expressed all expected cell surface markers, could differentiate to embryonic bodies upon culture in vitro, and differentiated further to derivatives of all three germ layers. CONCLUSION: This study indicates that homologous human fibroblasts can be used as feeder cells to support not only the propagation, but also the derivation of ES cells, and this should facilitate studies of therapeutic cloning for research and clinical applications.


Asunto(s)
Línea Celular , Células Madre Embrionarias , Prepucio/citología , Técnicas de Cultivo de Célula , Técnicas de Cocultivo , Fibroblastos/citología , Humanos , Cariotipificación , Masculino
20.
Zhonghua Yi Xue Za Zhi ; 90(3): 208-12, 2010 Jan 19.
Artículo en Zh | MEDLINE | ID: mdl-20356560

RESUMEN

OBJECTIVE: To construct the human aquaporin-4 (AQP4) expressing vector and detect anti-AQP4 antibody in serum of patients with neuromyelitis optica (NMO). METHODS: RNA was extracted from human glioblastoma and AQP4 cDNA obtained through RT-PCR.The fragment was cloned into the lentiviral expressing vector (iDUET101) and transformed into competent strain Hb101 for later amplification; plasmids were extracted from the amplified positive-bacteria-colony, sequenced and transfected into HEK-293T cells. Expression of AQP4 was identified by RT-PCR, Western blot and immunofluorescence assay. And anti-AQP4 antibody in human serum was tested. RESULTS: The sequence of target fragment matched with that of human AQP4 fragment sequences (NM_001650) completely. The constructed AQP4 fragment transfected in HEK-293T cell was tested by immunofluorescent examination and it exhibited obvious fluorescence located in cell membrane. Western blot test was positive. And the fragment was about 34 KD. Cellular immunofluorescence examination showed 11 examples of 12 NMO patient serums (91.7%) were positive, 4 in 34 multiple sclerosis (11.8%) positive and negative in all 50 serum samples of healthy controls. CONCLUSION: The HEK-293T cell transfected with lentivirus-AQP4 vector can express stably. And the expressed fragment may be applied in clinical examination.


Asunto(s)
Acuaporina 4/genética , Acuaporina 4/inmunología , Vectores Genéticos/biosíntesis , Neuromielitis Óptica/diagnóstico , Autoanticuerpos/sangre , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoglobulina G/sangre , Lentivirus/genética , Lentivirus/inmunología , Esclerosis Múltiple/diagnóstico , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA