Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Neurol Neurosurg ; 210: 106963, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34715556

RESUMEN

OBJECTIVES: This study aimed to analyze the difference between cerebral salt-wasting syndrome (CSWS) and syndrome of inappropriate secretion of antidiuretic hormone (SIADH) in patients with hyponatremia after hypothalamic tumor surgery, and to explore a convenient and effective way to identify CSWS and SIADH. METHODS: Patients undergoing craniotomy of hypothalamic tumor admitted to the Department of The Affiliated Hospital of Qingdao University from December 2018 to May 2020 were enrolled in this study. Plasma brain natriuretic peptide (BNP), 24-h urine sodium, 24-h urine volume, and the diameter of the inferior vena cava (IVCD) were measured daily before operation and 1-7 days after operation, to analyze differences in plasma BNP, 24-h urinary sodium excretion, 24-h urine volume, and IVCD between the CSWS and SIADH. RESULTS: The medical data of 31 patients with hypothalamic tumors were collected. Fifteen of these patients (48%) had postoperative hyponatremia, nine patients (29%) had CSWS, and six patients (19%) had SIADH. Plasma BNP, 24-h urinary sodium excretion, and 24-h urine volume in the CSWS group were significantly higher than those in the SIADH group. IVCD decreased in the CSWS group and increased in the SIADH group. CONCLUSIONS: When hyponatremia occurs after hypothalamic tumor surgery, plasma BNP, 24-h urinary sodium excretion, 24-h urine volume, and IVCD are of great help in identifying CSWS and SIADH.


Asunto(s)
Craneotomía/efectos adversos , Hiponatremia/etiología , Neoplasias Hipotalámicas/cirugía , Hipotálamo/cirugía , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/etiología
2.
Aging (Albany NY) ; 13(2): 3060-3079, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479185

RESUMEN

To investigate the therapeutic mechanism of action of transplanted stem cells and develop exosome-based nanotherapeutics for ischemic stroke, we assessed the effect of exosomes (Exos) produced by human umbilical cord mesenchymal stem cells (hUMSCs) on microglia-mediated neuroinflammation after ischemic stroke. Our results found that injected hUMSC-Exos were able to access the site of ischemic damage and could be internalized by cells both in vivo and in vitro. In vitro, treatment with hUMSC-Exos attenuated microglia-mediated inflammation after oxygen-glucose deprivation (OGD). In vivo results demonstrated that treatment with hUMSC-Exos significantly reduced infarct volume, attenuated behavioral deficits, and ameliorated microglia activation, as measured three days post-transient brain ischemia. Furthermore, miR-146a-5p knockdown (miR-146a-5p k/d Exos) partially reversed the neuroprotective effect of hUMSC-Exos. Our mechanistic study demonstrated that miR-146a-5p in hUMSC-Exos reduces microglial-mediated neuroinflammatory response through IRAK1/TRAF6 pathway. We conclude that miR-146a-5p derived from hUMSC-Exos can attenuate microglia-mediated neuroinflammation and consequent neural deficits following ischemic stroke. These results elucidate a potential therapeutic mechanism of action of mesenchymal stem cells and provide evidence that hUMSC-Exos represent a potential cell-free therapeutic option for ischemic stroke.


Asunto(s)
Exosomas/metabolismo , Inflamación/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Cordón Umbilical/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Transducción de Señal/fisiología
3.
J Craniofac Surg ; 32(1): 228-232, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33186284

RESUMEN

PURPOSE: To evaluate various factors that could be associated with the postoperative prognosis of patients with craniopharyngiomas and provide evidence for the proper surgical course and optimal outcome assessments of craniopharyngiomas. METHODS: We performed a retrospective study and reviewed 68 patients with craniopharyngiomas who received surgery from May 2013 to October 2018. The relationships between the disease prognosis and age, gender, onset symptoms, size of tumor, degree of calcification, consistency, QST classification, adhesion strength, and pathological types were analyzed. RESULTS: There were no significant associations between the prognosis and age, gender, number of onset symptoms, and pathological types (P > 0.05). The severity of onset symptoms, tumor diameter, and degree of calcification was significantly associated with the prognosis (P < 0.05). There were significant different prognoses between patients with cystic and solid, mixed tumors (P < 0.05). The prognosis of patients with T type tumors was different from that of patients with either Q or S type tumors (P < 0.05). The prognoses of patients with either loose or tight type tumors were significantly different from those of patients with either invasive or fusion type tumors (P < 0.05). CONCLUSION: Clinical and pathological variables, such as onset symptoms, size of tumor, degree of calcification, consistency, QST classification, and the degree of adhesion strength, were important factors in evaluating the prognosis of patients with craniopharyngiomas.


Asunto(s)
Craneofaringioma/cirugía , Neoplasias Hipofisarias/cirugía , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Periodo Posoperatorio , Pronóstico , Estudios Retrospectivos , Adulto Joven
4.
Cell Cycle ; 19(24): 3468-3479, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33269647

RESUMEN

Store-operated Ca2+ entry (SOCE) plays an important role in regulating Ca2+ influx, which participates in tumor cell survival and motility. We aim to elucidate the role of SOCE in the behavior of C6 glioma cells. Lentiviral vector inserted with the Orai1-targeting shRNA was used to inhibit SOCE in C6 glioma cells. The down-regulation of Orai1 was confirmed by western blot. The ability of shOrai1 or SOCE inhibitor (SKF96365) in regulating SOCE inhibition was evaluated by measuring Ca2+ concentration. Additionally, its effect on cell behavior was assessed using methyl thiazolyl tetrazolium (MTT) assay, wound healing assay, transwell assay, and adhesion assay. Focal adhesions were visualized by immunofluorescence assay. Further, the expression of proline-rich tyrosine kinase 2 (Pyk2) and phosphorylated Pyk2 (p-Pyk2) was analyzed using western blot. Both, SKF96365 treatment and the Orai1 down-regulation inhibited SOCE by perturbing Ca2+ influx. The inhibitory effects of shOrai1 on C6 cell proliferation, migration, and invasion were similar to that of SKF96365. Moreover, Orai1 inhibition enhanced C6 cell adhesion by increasing the size of focal adhesion plaques. The down-regulation of Pyk2 was observed in both SKF96365-treated and Orai1-silenced C6 cells. Additionally, Orai1 inhibition blocked AKT/mTOR, NFAT, and NF-κB pathways. The silencing of Orai1 inhibited the C6 glioma cell migration, invasion and contributed to focal adhesion.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Señalización del Calcio/genética , Calcio/metabolismo , Movimiento Celular/genética , Quinasa 2 de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Glioma/metabolismo , Proteína ORAI1/genética , Interferencia de ARN , Animales , Neoplasias Encefálicas/patología , Señalización del Calcio/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Regulación hacia Abajo/genética , Adhesiones Focales/genética , Glioma/patología , Imidazoles/farmacología , Invasividad Neoplásica/genética , Proteína ORAI1/metabolismo , Ratas
5.
World Neurosurg ; 139: 223-225, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32305611

RESUMEN

BACKGROUND: Osteosarcoma is a common malignant bone tumor that occurs in children or adolescents but rarely in the skull. Epidermoid cysts, also known as cholesteatomas, represent approximately 0.2%-1.8% of all intracranial tumors. The occurrence of osteosarcoma with an epidermoid cyst is extremely rare. CASE DESCRIPTION: A 41-year-old woman had both osteosarcoma and cholesteatoma in the left cerebellopontine angle. We resected the 2 tumors using the suboccipital retrosigmoid approach, and she received radiotherapy and chemotherapy after the surgery. One year after surgery, the patient is healthy and has recovered well. CONCLUSIONS: Osteosarcomas and epidermoid cysts should be completely resected to prevent tumor recurrence and aseptic meningitis. Postoperative osteosarcoma treatment should include radiotherapy and chemotherapy to improve the survival rate of patients. It is hoped that this report will help clinicians in diagnosis and treatment of patients with similar conditions.


Asunto(s)
Encefalopatías/patología , Neoplasias Cerebelosas/patología , Ángulo Pontocerebeloso/patología , Colesteatoma/patología , Osteosarcoma/patología , Adulto , Encefalopatías/complicaciones , Neoplasias Cerebelosas/complicaciones , Colesteatoma/complicaciones , Quiste Epidérmico/complicaciones , Quiste Epidérmico/patología , Femenino , Humanos , Osteosarcoma/complicaciones
6.
Exp Neurol ; 313: 49-59, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30213507

RESUMEN

Bone marrow-derived mesenchymal stem cells (BMSCs) exhibit potential regenerative effects on the injured brain. However, these effects are constrained by their limited ability to migrate to the injured site. Oncostatin M (OSM) has been shown to affect the proliferation and migration of mesenchymal stem cells. Therefore, in the present study, we explored whether OSM improves BMSC migration and secretion of growth factors and cytokines in a rat middle cerebral artery occlusion (MCAO) stroke model. The effect of OSM on the proliferation and apoptosis of rat BMSCs was first assessed in vitro, and the gene and secretion levels of factors related to cell nutrition and migration, such as SDF-1 and VEGF, were detected. To further explore underlying pathways triggered by OSM, BMSCs were treated with OSM in the presence or absence of inhibitors of the STAT3 and ERK pathways. Effects of OSM on SDF-1 expression in astrocytes and BMSC migration were also evaluated. In the rat MCAO model, OSM secretion levels were detected in the brain for up to 72 h after model establishment. Ventricle injection of OSM alone or OSM combined with caudal vein graft of BMSCs was then performed in MCAO stroke rats. After 72 h, production of SDF-1 and grafted BMSCs was detected in the lesion areas of the brain, and the nerve function score was evaluated. We found that the production of OSM continually increased in the brains of MCAO rats from 12 h to 72 h. OSM significantly upregulated SDF-1 in BMSCs via the STAT3 and ERK pathways and significantly promoted the expression of VEGF and MMP-2. OSM also promoted the secretion of SDF-1 in astrocytes through the STAT3 and ERK pathways to in turn enhance BMSC migration. Combination treatment with OSM and BMSCs in MCAO rats increased the migration efficiency of BMSCs in the brain, which significantly improved neurofunctional recovery while reducing the expression of inflammatory mediators and promoting the secretion of nutrition factors. Overall, these results show that OSM is highly expressed in the brains of MCAO stroke rats and can upregulate SDF-1 to promote BMSC migration. Thus, combination treatment with OSM and BMSCs improves the graft efficiency of BMSCs and neurofunctional recovery.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/biosíntesis , Inhibidores de Crecimiento/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Oncostatina M/farmacología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Inhibidores de Crecimiento/metabolismo , Inhibidores de Crecimiento/uso terapéutico , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Trasplante de Células Madre Mesenquimatosas , Oncostatina M/metabolismo , Oncostatina M/uso terapéutico , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Factor de Transcripción STAT3/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Regulación hacia Arriba/efectos de los fármacos
7.
Oncoimmunology ; 7(2): e1391973, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29308321

RESUMEN

Tumor initiating cells or cancer stem cells (CSCs) play an important role in the initiation, development, metastasis, and recurrence of tumors. However, traditional therapies have limited effects against CSCs and targeting these cells is crucial when developing new therapeutic strategies against cancer. One potentially targetable factor is CD47, a member of the immunoglobulin superfamily. This protein acts as an anti-phagocytic "don't eat me" signal and is often found expressed by cancer cells, particularly CSCs. CD47 functions by activating signal regulatory protein-α (SIRP-α) expressed on macrophages, preventing phagocytosis. However, the role of CD47 in glioma stem cells (GSCs) has been not been thoroughly investigated. Our study therefore examined the expression and function of this protein in glioma cells and GSCs. We found that CD47 was highly expressed on glioma cells, especially GSCs, and that expression associated with worse clinical outcomes. We also found that CD47+ glioma cells possessed stem/progenitor cell-like characteristics and knocking down CD47 expression resulted in a reduction in these characteristics. Treatment with anti-CD47 antibody led to increased phagocytosis of glioma cells and GSCs by macrophages. We next examined the effects of anti-CD47 antibody on glioma cells/GSCs in an immune competent mouse glioma model, revealing significant inhibition of tumor growth and prolonged survival times. Importantly, there were no apparent side effects in the animal model. In summary, we have shown that CD47 is a potentially safe and effective therapeutic target for glioma.

8.
Am J Transl Res ; 9(5): 2492-2499, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28559999

RESUMEN

Mesenchymal stem cell (MSC) transplantation is a promising therapeutic strategy for myocardial infarction. The survival rate of the grafted MSCs is limited by the conditions of hypoxia and low nutrient levels. In this study, we investigated the role of hypoxia-inducible factor 1 alpha (Hif-1α) in oxygen-glucose deprivation (OGD)-induced injury in MSCs. Hif-1α was overexpressed or suppressed in MSCs by transfection with a Hif-1α expressing vector or Hif-1α-specific siRNA, respectively. Then MSCs were exposed to OGD, and the changes in cell viability, cell cycle distribution and apoptosis were respectively monitored by MTT assay and flow cytometry. Additionally, expression levels of Beclin1, LC3 I and LC3 II, as well phosphorylation of PI3K, AKT and mTOR were detected by RT-PCR and Western blotting. The results indicated that Hif-1α overexpression improved cell viability, reduced G1 phase cells accumulation, and suppressed apoptosis under OGD condition (P<0.05). Beclin1 expression and the LC3 II/LC3 I ratio were increased by Hif-1α overexpression, and were decreased by Hif-1α knock-down (P<0.05). In addition, PI3K, AKT and mTOR were inactivated by Hif-1α overexpression, and phosphorylated by Hif-1α knock-down (P<0.05). In conclusion, these data suggest that Hif-1α overexpression protects MSCs from OGD-induced injury via a mechanism in which autophagy and PI3K/AKT/mTOR pathway are implicated.

9.
Am J Transl Res ; 9(3): 1521-1529, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28386377

RESUMEN

Mesenchymal stem cells (MSCs) are ideal materials for cell therapy. Research has indicated that hypoxia benefits MSC survival, but little is known about the underlying mechanism. This study aims to uncover potential mechanisms involving hypoxia inducible factor 1α (HIF1A) to explain the promoted MSC survival under hypoxia. MSCs were obtained from Sprague-Dawley rats and cultured under normoxia or hypoxia condition. The overexpression vector or small interfering RNA of Hif1a gene was transfected to MSCs, after which cell viability, apoptosis and expression of HIF1A were analyzed by MTT assay, flow cytometry, qRT-PCR and Western blot. Factors in p53 pathway were detected to reveal the related mechanisms. Results showed that hypoxia elevated MSCs viability and up-regulated HIF1A (P < 0.05) as previously reported. HIF1A overexpression promoted viability (P < 0.01) and suppressed apoptosis (P < 0.001) under normoxia. Correspondingly, HIF1A knockdown inhibited viability (P < 0.05) and promoted apoptosis (P < 0.01) of MSCs under hypoxia. Expression analysis suggested that p53, phosphate-p53 and p21 were repressed by HIF1A overexpression and promoted by HIF1A knockdown, and B-cell CLL/lymphoma 2 (BCL2) expression had the opposite pattern (P < 0.05). These results suggest that HIF1A may improve viability and suppress apoptosis of MSCs, implying the protective effect of HIF1A on MSC survival under hypoxia. The underlying mechanisms may involve the HIF1A-suppressed p53 pathway. This study helps to explain the mechanism of MSC survival under hypoxia, and facilitates the application of MSCs in cell therapy.

10.
Front Mol Neurosci ; 10: 80, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28424584

RESUMEN

Bone mesenchymal stem cells (BMSCs) death after transplantation is a serious obstacle impacting on the outcome of cell therapy for cerebral infarction. This study was aimed to investigate whether modification of BMSCs with hypoxia-inducible factor 1α (Hif-1α) could enhance the survival of the implanted BMSCs. BMSCs were isolated from Wistar rats, and were infected with Hif-1α-GFP lentiviral vector or Hif-1α siRNA. The modified BMSCs were exposed to oxygen-glucose deprivation (OGD) condition, cellular viability and apoptosis were then assessed. An inhibitor of AMPK (compound C) was used to detect whether AMPK and mTOR were implicated in the functions of Hif-1α on BMSCs survival. Besides, ultrastructure of BMSCs was observed and the expression of autophagy markers was measured. The modified BMSCs were transplanted into middle cerebral artery occlusion (MCAO) model of rats, and the cerebral infarction volume and neurological function was assessed. The results indicated that Hif-1α overexpression protected OGD induced injury by promoting cellular viability and inhibiting apoptosis. AMPK was activated while mTOR was inactivated by Hif-1α overexpression, and that might be through which Hif-1α functioned BMSCs survival. Hif-1α overexpression promoted autophagy; more important, compound C abolished the induction of Hif-1α on autophagy. Transplantation of the overexpressed Hif-1α of BMSCs into the MCAO rats reduced brain infarct volume and improved neurobehavioral outcome; besides, it inhibited pro-inflammatory cytokines generation while promoted neurotrophin secretion. In conclusion, Hif-1α might be contributed in the survival of BMSCs by regulating the activation of AMPK and mTOR, as well as by promoting autophagy.

11.
Front Cell Neurosci ; 10: 283, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28018176

RESUMEN

In this study, we investigated interactions among microglia (MG), bone marrow mesenchymal stem cells (BMSCs) and neurons in cerebral ischemia and the potential mechanisms using an in vitro oxygen-glucose deprivation (OGD) model. Rat BMSCs were incubated with conditioned medium (CM) from in vitro cultures of OGD-activated rat MG and murine BV2 MG cells. Effects of glial cell-derived neurotrophic factor (GDNF) on rat neuron viability, apoptosis, lactate dehydrogenase (LDH) leakage and mitochondrial membrane potential (MMP) were analyzed in this model. OGD-activated MG promoted GDNF production by BMSCs (P < 0.01). Tumor necrosis factor-α (TNFα), but not interleukin-6 (IL6) or interleukin 1ß (IL1ß), promoted GDNF production by BMSCs (P < 0.001). GDNF or CM pre-treated BMSCs elevated neuronal viability and suppressed apoptosis (P < 0.05 or P < 0.01); these effects were inhibited by the RET antibody. GDNF activated MEK/ERK and phosphoinositide-3-kinase (PI3K)/AKT signaling but not JNK/c-JUN. Furthermore, GDNF upregulated B cell lymphoma 2 (BCL2) and heat shock 60 kDa protein 1 (HSP60) levels, suppressed LDH leakage, and promoted MMP. Thus, activated MG produce TNFα to stimulate GDNF production by BMSCs, which prevents and repairs OGD-induced neuronal injury, possibly via regulating MEK/ERK and PI3K/AKT signaling. These findings will facilitate the prevention and treatment of neuronal injury by cerebral ischemia.

12.
J Cell Physiol ; 230(10): 2461-75, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25752945

RESUMEN

Astrocytes are critical for ischemic stroke, and understanding their role in mesenchymal stem cell (MSC)-mediated protection against ischemic injury is important. The paracrine capacity of MSCs has been proposed as the principal mechanism contributing to the protection and repair of brain tissue. In the present study, an in vitro oxygen-glucose deprivation (OGD) model was used to mimic ischemic injury. OGD-induced astrocytes were reperfused with MSC-conditioned medium (MSC-CM) or co-cultured with MSCs for 24 h to create an environment abundant in paracrine factors. The results indicated that both situations could protect astrocytes from apoptosis, increase cell metabolic activity, and reduce glial fibrillary acidic protein (GFAP) overexpression; however, the effects of co-culturing with MSCs were more positive. Paracrine factors suppressed the activation of p38 MAPK, JNK, and their downstream targets p53 and STAT1. Inhibition of p38 MAPK, JNK, p53, and STAT1 attenuated astrocyte injury and/or GFAP upregulation. Activation of p38 MAPK and JNK suppressed the beneficial effects of paracrine factors, resulting in decreased survival and GFAP overexpression. These results suggest that paracrine factors inhibit p38 MAPK and JNK, and most likely by regulating their downstream targets, p53 and STAT1, to promote astrocyte survival associated with GFAP downregulation after ischemic stroke in vitro.


Asunto(s)
Astrocitos/metabolismo , Isquemia Encefálica/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Células Madre Mesenquimatosas/metabolismo , Accidente Cerebrovascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Comunicación Paracrina , Ratas Wistar
13.
J Neuroinflammation ; 11: 135, 2014 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-25088370

RESUMEN

Microglia are the primary immunocompetent cells in brain tissue and microglia-mediated inflammation is associated with the pathogenesis of various neuronal disorders. Recently, many studies have shown that mesenchymal stem cells (MSCs) display a remarkable ability to modulate inflammatory and immune responses through the release of a variety of bioactive molecules, thereby protecting the central nervous system. Previously, we reported that MSCs have the ability to modulate inflammatory responses in a traumatic brain injury model and that the potential mechanisms may be partially attributed to upregulated TNF-α stimulated gene/protein 6 (TSG-6) expression. However, whether TSG-6 exerts an anti-inflammatory effect by affecting microglia is not fully understood. In this study, we investigated the anti-inflammatory effects of MSCs and TSG-6 in an in vitro lipopolysaccharide (LPS)-induced BV2 microglial activation model. We found that MSCs and TSG-6 significantly inhibited the expression of pro-inflammatory mediators in activated microglia. However, MSC effects on microglia were attenuated when TSG-6 expression was silenced. In addition, we found that the activation of nuclear factor (NF)-κB and mitogen-activated protein kinase (MAPK) pathways in LPS-stimulated BV2 microglial cells was significantly inhibited by TSG-6. Furthermore, we found that the presence of CD44 in BV2 microglial cells was essential for MSC- and TSG-6-mediated inhibition of pro-inflammatory gene expression and of NF-κB and MAPK activation in BV2 microglial cells. The results of this study suggest that MSCs can modulate microglia activation through TSG-6 and that TSG-6 attenuates the inflammatory cascade in activated microglia. Our study indicates that novel mechanisms are responsible for the immunomodulatory effect of MSCs on microglia and that MSCs, as well as TSG-6, might be promising therapeutic agents for the treatment of neurotraumatic injuries or neuroinflammatory diseases associated with microglial activation.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/fisiología , Microglía/efectos de los fármacos , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Moléculas de Adhesión Celular/genética , Línea Celular Transformada , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Antígenos de Histocompatibilidad Clase II/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Transfección , Factor de Necrosis Tumoral alfa/farmacología
14.
Biochem Biophys Res Commun ; 450(4): 1409-15, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25014173

RESUMEN

Dendritic cells (DCs) are potent antigen-presenting cells (APCs) that are characterized by the ability to take up and process antigens and prime T cell responses. Mesenchymal stem cells (MSCs) are multipotent cells that have been shown to have immunomodulatory abilities, including inhibition of DC maturation and function in vivo and in vitro; however, the underlying mechanism is far from clear. In this study we found that MSCs can inhibit the maturation and function of bone marrow-derived DCs by releasing TSG-6. In the presence of MSCs, lower expression of mature DC surface phenotype (CD80, CD86, MHC-II, and CD11c) was observed. In addition, typical DC functions, such as the production of IL-12 and the ability to prime T cells, were decreased when co-cultured with MSCs. In contrast, knockdown of TSG-6 reduced the inhibitory effect of MSCs on DC. Moreover, we found that TSG-6 can suppress the activation of MAPKs, and NF-κB signaling pathways within DCs during Lipopolysaccharides (LPS) stimulation. In conclusion, we suggest that TSG-6 plays an important role in MSCs-mediated immunosuppressive effect on DC.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/citología , Células Madre Mesenquimatosas/citología , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Interleucina-12/metabolismo , Activación de Linfocitos , Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores
15.
Brain Res ; 1590: 1-9, 2014 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-24909791

RESUMEN

Human amniotic membrane-derived mesenchymal stem cells (AMSCs) are considered a novel and promising source of stem cells for cell replacement-based therapy. Current research is mostly limited to investigating the cellular differentiation potential of AMSCs, while few have focused on their immunosuppressive properties. This study is aimed at exploring and evaluating the immunosuppressive effect of human AMSCs on the viability and migratory properties of microglia. We found, from results of cell viability assays, that AMSCs can reduce the activity of inflammatory cells by secreting nitric oxide (NO). Also, based on results from wound healing and transwell migration assays, we show that AMSCs can inhibit the migration of human microglia as well as the mouse microglial cell line BV2, suggesting that they have the ability to inhibit the recruitment of certain immune cells to injury sites. Furthermore, we found that NO contributes significantly to this inhibitory effect. Our study provides evidence that human AMSCs can have detrimental effects on the viability and migration of microglia, through secretion of NO. This mechanism may contribute to anti-inflammatory processes in the central nervous system.


Asunto(s)
Amnios/citología , Tolerancia Inmunológica/fisiología , Trasplante de Células Madre Mesenquimatosas , Microglía/fisiología , Óxido Nítrico/fisiología , Amnios/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Medios de Cultivo Condicionados , Humanos , Ratones , Óxido Nítrico/metabolismo , Reclutamiento Neurofisiológico , Cicatrización de Heridas
16.
PLoS One ; 8(12): e84116, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391898

RESUMEN

Many studies have shown that microglia in the activated state may be neurotoxic. It has been proven that uncontrolled or over-activated microglia play an important role in many neurodegenerative disorders. Bone marrow-derived mesenchymal stem cells (BMSCs) have been shown in many animal models to have a therapeutic effect on neural damage. Such a therapeutic effect is attributed to the fact that BMSCs have the ability to differentiate into neurons and to produce trophic factors, but there is little information available in the literature concerning whether BMSCs play a therapeutic role by affecting microglial activity. In this study, we triggered an inflammatory response situation in vitro by stimulating microglia with the bacterial endotoxin lipopolysaccharide (LPS), and then culturing these microglia with BMSC-conditioned medium (BMSC-CM). We found that BMSC-CM significantly inhibited proliferation and secretion of pro-inflammatory factors by activated microglia. Furthermore, we found that the phagocytic capacity of microglia was also inhibited by BMSC-CM. Finally, we investigated whether the induction of apoptosis and the production of nitric oxide (NO) were involved in the inhibition of microglial activation. We found that BMSC-CM significantly induced apoptosis of microglia, while no apoptosis was apparent in the LPS-stimulated microglia. Our study also provides evidence that NO participates in the inhibitory effect of BMSCs. Our experimental results provide evidence that BMSCs have the ability to maintain the resting phenotype of microglia or to control microglial activation through their production of several factors, indicating that BMSCs could be a promising therapeutic tool for treatment of diseases associated with microglial activation.


Asunto(s)
Médula Ósea/crecimiento & desarrollo , Proliferación Celular , Células Madre Mesenquimatosas/citología , Microglía/citología , Óxido Nítrico/metabolismo , Animales , Apoptosis , Médula Ósea/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Lipopolisacáridos/farmacología , Masculino , Células Madre Mesenquimatosas/metabolismo , Microglía/metabolismo , Fagocitosis/fisiología , Fenotipo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...