Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 7244, 2024 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-38538745

RESUMO

We aimed to evaluate whether white and gray matter microstructure changes observed with magnetic resonance imaging (MRI)-based diffusion tensor imaging (DTI) can be used to reflect the progression of chronic brain trauma. The MRI-DTI parameters, neuropathologic changes, and behavioral performance of adult male Wistar rats that underwent moderate (2.1 atm on day "0") or repeated mild (1.5 atm on days "0" and "2") traumatic brain injury (TBI or rmTBI) or sham operation were evaluated at 7 days, 14 days, and 1-9 months after surgery. Neurobehavioral tests showed that TBI causes long-term motor, cognitive and neurological deficits, whereas rmTBI results in more significant deficits in these paradigms. Both histology and MRI show that rmTBI causes more significant changes in brain lesion volumes than TBI. In vivo DTI further reveals that TBI and rmTBI cause persistent microstructural changes in white matter tracts (such as the body of the corpus callosum, splenium of corpus callus, internal capsule and/or angular bundle) of both two hemispheres. Luxol fast blue measurements reveal similar myelin loss (as well as reduction in white matter thickness) in ipsilateral and contralateral hemispheres as observed by DTI analysis in injured rats. These data indicate that the disintegration of microstructural changes in white and gray matter parameters analyzed by MRI-DTI can serve as noninvasive and reliable markers of structural and functional level alterations in chronic TBI.


Assuntos
Lesões Encefálicas Traumáticas , Substância Branca , Masculino , Ratos , Animais , Imagem de Tensor de Difusão/métodos , Substância Cinzenta/diagnóstico por imagem , Substância Cinzenta/patologia , Ratos Wistar , Imageamento por Ressonância Magnética , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Lesões Encefálicas Traumáticas/patologia , Substância Branca/diagnóstico por imagem , Substância Branca/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia
2.
Sci Prog ; 107(1): 368504241231154, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38425276

RESUMO

The underlying mechanisms for the beneficial effects exerted by bone marrow-mesenchymal stem cells (BM-MSCs) in treating repetitive traumatic brain injury (rTBI)-induced long-term sensorimotor/cognitive impairments are not fully elucidated. Herein, we aimed to explore whether BM-MSCs therapy protects against rTBI-induced long-term neurobehavioral disorders in rats via normalizing white matter integrity and gray matter microglial response. Rats were subjected to repeated mild lateral fluid percussion on day 0 and day 3. On the fourth day post-surgery, MSCs groups received MSCs (4 × 106 cells/ml/kg, intravenously) and were assessed by the radial maze, Y maze, passive avoidance tests, and modified neurological severity scores. Hematoxylin & eosin, and Luxol fast blue stainings were used to examine the histopathology and white matter thickness. At the same time, immunofluorescence staining was used to investigate the numbers of tumor necrosis factor-alpha (TNF-α)-containing microglia in gray matter. Three to nine months after neurotrauma, rats displayed sensorimotor and cognitive impairments, reduced thickness in white matter, and over-accumulation of TNF-α-containing microglia and cellular damage in gray matter. Therapy with BM-MSCs significantly attenuated the rTBI-induced sensorimotor and cognitive impairments and all their complications. Mesenchymal stem cell therapy might accelerate the recovery of sensorimotor and cognitive impairments in rats with rTBI via normalizing myelin integrity and microglia response.


Assuntos
Lesões Encefálicas Traumáticas , Disfunção Cognitiva , Células-Tronco Mesenquimais , Ratos , Animais , Bainha de Mielina , Microglia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia , Lesões Encefálicas Traumáticas/terapia , Cognição
3.
Aging Cell ; 20(5): e13340, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33783931

RESUMO

Amyloid-beta (Aß) oligomer is known to contribute to the pathophysiology of age-related macular degeneration. Herein, we aimed to elucidate the in vivo and in vitro effects of Aß1-42 application on retinal morphology in rats. Our in vivo studies revealed that intracerebroventricular administration of Aß1-42 oligomer caused dysmorphological changes in both retinal ganglion cells and retinal pigment epithelium. In addition, in vitro studies revealed that ARPE-19 cells following Aß1-42 oligomer application had decreased viability along with apoptosis and decreased expression of the tight junction proteins, increased expression of both phosphor-AKT and phosphor-GSK3ß and decreased expression of both SIRT1 and ß-catenin. Application of conditioned medium (CM) obtained from mesenchymal stem cells (MSC) protected against Aß1-42 oligomer-induced retinal pathology in both rats and ARPE-19 cells. In order to explore the potential role of peptides secreted from the MSCs, we applied mass spectrometry to compare the peptidomics profiles of the MSC-CM. Gene ontology enrichment analysis and String analysis were performed to explore the differentially expressed peptides by predicting the functions of their precursor proteins. Bioinformatics analysis showed that 3-8 out of 155-163 proteins in the MSC-CM maybe associated with SIRT1/pAKT/pGSK3ß/ß-catenin, tight junction proteins, and apoptosis pathway. In particular, the secretomes information on the MSC-CM may be helpful for the prevention and treatment of retinal pathology in age-related macular degeneration.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Células-Tronco Mesenquimais/metabolismo , Retina/patologia , Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides , Animais , Apoptose , Hipóxia Celular , Linhagem Celular , Meios de Cultivo Condicionados , Modelos Animais de Doenças , Humanos , Aprendizagem , Fragmentos de Peptídeos , Ratos , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Transdução de Sinais , Memória Espacial , Proteínas de Junções Íntimas/metabolismo , beta Catenina/metabolismo
4.
Sci Rep ; 10(1): 14409, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32873851

RESUMO

To date, there is no good evidence that intestine epithelial cells (IEC) affected by ischemia/reperfusion (I/R) injury are able to cause cortical neuron injury directly. Additionally, it remains unclear whether the neuronal damage caused by I/R injured IEC can be affected by therapeutic hypothermia (TH, 32 °C). To address these questions, we performed an oxygen-glucose deprivation (OGD) affected IEC-6-primary cortical neuron coculture system under normothermia (37 °C) or TH (32 °C) conditions. It was found that OGD caused hyperpermeability in IEC-6 cell monolayers. OGD-preconditioned IEC-6 cells caused cortical neuronal death (e.g., decreased cell viability), synaptotoxicity, and neuronal apoptosis (evidenced by increased caspase-3 expression and the number of TUNEL-positive cells), necroptosis (evidenced by increased receptor-interacting serine/threonine-protein kinase-1 [RIPK1], RIPK3 and mixed lineage kinase domain-like pseudokinase [MLKL] expression), and pyroptosis (evidenced by an increase in caspase-1, gasdermin D [GSDMD], IL-1ß, IL-18, the apoptosis-associated speck-like protein containing a caspase recruitment domain [ASC], and nucleotide oligomerization domain [NOD]-like receptor [NLRP]-1 expression). TH did not affect the intestinal epithelial hyperpermeability but did attenuate OGD-induced neuronal death and synaptotoxicity. We also performed quantitative real-time PCR to quantify the genes encoding 84 exosomal microRNAs in the medium of the control-IEC-6, the control-neuron, the OGD-IEC-6 at 37 °C, the OGD-IEC-6 at 32 °C, the neuron cocultured with OGD-IEC-6 at 37 °C, and the neurons cocultured with OGD-IEC-6 at 32 °C. We found that the control IEC-6 cell s or cortical neurons are able to secrete a basal level of exosomal miRNAs in their medium. OGD significantly up-regulated the basal level of each parameter for IEC-6 cells. As compared to those of the OGD-IEC-6 cells or the control neurons, the OGD-IEC-6 cocultured neurons had significantly higher levels of 19 exosomal miRNAs related to apoptosis, necroptosis, and/or pyroptosis events. Our results identify that I/R injured intestinal epithelium cells can induce cortical neuron death via releasing paracrine mediators such as exosomal miRNAs associated with apoptosis, necroptosis, and/or pyroptosis, which can be counteracted by TH.


Assuntos
Hipóxia Celular , Córtex Cerebral/citologia , Células Epiteliais/metabolismo , Exossomos/metabolismo , Glucose/metabolismo , Mucosa Intestinal/citologia , MicroRNAs/metabolismo , Necroptose , Neurônios/metabolismo , Piroptose , Animais , Linhagem Celular , Sobrevivência Celular , Córtex Cerebral/embriologia , Técnicas de Cocultura , Hipotermia/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo
5.
Med Sci Monit ; 24: 8096-8104, 2018 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-30417859

RESUMO

BACKGROUND Several clinical conditions can cause hepatic ischemia/reperfusion (I/R) injury. This study aimed to determine the mechanism of the protective effect of hyperbaric oxygen preconditioning (HBO2P) on hepatic ischemia/reperfusion (I/R) injury in a rat model, and to investigate the effects on HBO2P and I/R injury of blocking HSP70 using antibody (Ab) pretreatment. MATERIAL AND METHODS Male Sprague-Dawley rats underwent HBO2P for 60 min at 2.0 atmosphere absolute (ATA) pressure for five consecutive days before surgical hepatic I/R injury, performed by clamping the portal vein and hepatic lobe. Four groups studied included: the non-HBO2P+ non-I/R group, which underwent sham surgery (N=10); the non-HBO2P + I/R group (N=10); the HBO2P + I/R group (N=10); and the HBO2P + HSP70-Ab + I/R group (N=10) received one dose of HSP70 antibody one day before hepatic I/R injury. Serum lactate dehydrogenase (LDH), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), and hepatic malondialdehyde (MDA) and myeloperoxidase (MPO) were measured biochemically. Rat liver tissues were examined histologically. RESULTS In rats with hepatic I/R injury without HSP70 antibody pre-treatment, HBO2P significantly reduced hepatic injury and levels of LDH, AST, ALT, TNF-α, IL-6, MDA, and MPO levels; in comparison, the group pre-treated with an antibody to inhibit HSP70 (the HBO2P + HSP70-Ab + I/R group) showed significant reversal of the beneficial effects of HBO2P on hepatic I/R injury (p<0.05). CONCLUSIONS In a rat model of hepatic I/R injury with HBO2P, HSP70 reduced hepatic inflammatory and oxidative damage.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Oxigenoterapia Hiperbárica/métodos , Fígado/irrigação sanguínea , Traumatismo por Reperfusão/prevenção & controle , Animais , Aspartato Aminotransferases/sangue , Modelos Animais de Doenças , Imuno-Histoquímica , Interleucina-6/sangue , L-Lactato Desidrogenase/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Hepatopatias/prevenção & controle , Masculino , Estresse Oxidativo/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Fator de Necrose Tumoral alfa/sangue
6.
Oncotarget ; 9(2): 1992-2001, 2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29416747

RESUMO

CD34 is a transmembrane phosphoglycoprotein used to selectively enrich bone marrow in hematopoietic stem cells for transplantation. Treating rats with CD34+ cells derived from human umbilical cord blood before or after heat stroke has been shown to promote survival. We investigated whether CD34- human placenta-derived stem cells (PDMSCs) could improve survival following heat stroke in rats. Rats were subjected to heat stress (42°C for 98 min) to induce heat stroke. Intravenous administration of PDMSCs 1 day before or immediately after the onset of heat stroke improved survival by 60% and 20%, respectively. Pre-treatment with CD34- PDMSCs protected against heat stroke injury more effectively than that treatment after injury. PDMSCs treatment attenuated cerebrovascular dysfunction, the inflammatory response, and lipid peroxidation. These data suggest human PDMSCs protect against heat stroke injury in rats. Moreover, these effects do not require the presence of CD34+ cells.

7.
J Pharm Pharmacol ; 70(6): 760-767, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29484657

RESUMO

OBJECTIVES: This study aimed to investigate the cardioprotective effects of melatonin on heat stroke (HS) induced acute myocardial infarction in rats and to explore the underlying mechanisms. METHODS: Myocardial injury was induced by subjecting the anaesthetized rats to a high ambient temperature of 43°C for 70 min. Such a high ambient temperature caused hyperthermia, hypotension and myocardial injury in rats. Rats were treated with melatonin (3 mg/kg) intravenously one hour before and followed by an additional dose immediately after heat stress. KEY FINDINGS: At the onset of HS, animals displayed myocardial injury evidenced by increased levels of cardiac damage indicators (e.g. total lactate dehydrogenase, cardiac troponin I and creatine kinase-MB), increased cardiac damage scores and suppressed left ventricular performance. Animals with HS also had increased cardiac oxidative stress evidenced by increased levels of lipid peroxidation (e.g. increased thiobarbituric acid reactive substances) and decreased levels of antioxidant enzymes (e.g. superoxide dismutase, catalase and reduced glutathione) and activated inflammation (e.g. increased levels of interleukin-6 and tumour necrosis factor-α). Pretreatment with melatonin significantly reversed the HS-induced myocardial injury, cardiac oxidative stress and cardiac inflammation. CONCLUSIONS: Melatonin may protect against HS-induced myocardial injury in male rats by mitigating oxidative stress and inflammation.


Assuntos
Golpe de Calor/complicações , Melatonina/farmacologia , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/prevenção & controle , Animais , Cardiotônicos/farmacologia , Inflamação/prevenção & controle , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos
8.
Int J Med Sci ; 14(13): 1327-1334, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29200946

RESUMO

Background: Clinical assessment reveals that patients after surgery of cardiopulmonary bypass or coronary bypass experience postoperative cognitive dysfunction. This study aimed to investigate whether resuscitation after a hemorrhagic shock (HS) and/or mild cerebral ischemia caused by a unilateral common carotid artery occlusion (UCCAO) can cause brain injury and concomitant neurological dysfunction, and explore the potential mechanisms. Methods: Blood withdrawal (6 mL/100 g body weight) for 60 min through the right jugular vein catheter-induced an HS. Immediately after the termination of HS, we reinfused the initially shed blood volumes to restore and maintain the mean arterial blood pressure (MABP) to the original value during the 30-min resuscitation. A cooling water blanket used to induce whole body cooling for 30 min after the end of resuscitation. Results: An UCCAO caused a slight cerebral ischemia (cerebral blood flow [CBF] 70%) without hypotension (MABP 85 mmHg), systemic inflammation, multiple organs injuries, or neurological injury. An HS caused a moderate cerebral ischemia (52% of the original CBF levels), a moderate hypotension (MABP downed to 22 mmHg), systemic inflammation, and peripheral organs injuries. However, combined an UCCAO and an HS caused a severe cerebral ischemia (18% of the original CBF levels), a moderate hypotension (MABP downed to 17 mmHg), systemic inflammation, peripheral organs damage, and neurological injury, which can be attenuated by whole body cooling. Conclusions: When combined with an HS, an UCCAO is associated with ischemic neuronal injury in the ipsilateral hemisphere of adult rat brain, which can be attenuated by therapeutic hypothermia. A resuscitation from an HS regards as a reperfusion insult which may induce neurological injury in patients with an UCCAO disease.


Assuntos
Lesões Encefálicas/fisiopatologia , Isquemia Encefálica/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Hipotensão/fisiopatologia , Animais , Pressão Sanguínea , Lesões Encefálicas/etiologia , Isquemia Encefálica/complicações , Ponte Cardiopulmonar/efeitos adversos , Artéria Carótida Primitiva/fisiopatologia , Artéria Carótida Primitiva/cirurgia , Circulação Cerebrovascular/fisiologia , Transtornos Cerebrovasculares/complicações , Transtornos Cerebrovasculares/fisiopatologia , Disfunção Cognitiva/etiologia , Modelos Animais de Doenças , Humanos , Hipotensão/etiologia , Complicações Pós-Operatórias , Ratos , Ressuscitação/efeitos adversos , Choque Hemorrágico/complicações , Choque Hemorrágico/fisiopatologia
9.
J Neuroinflammation ; 14(1): 90, 2017 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-28438174

RESUMO

BACKGROUND: Despite previous evidence for a potent inflammatory response after a traumatic brain injury (TBI), it is unknown whether exercise preconditioning (EP) improves outcomes after a TBI by modulating inflammatory responses. METHODS: We performed quantitative real-time PCR (qPCR) to quantify the genes encoding 84 cytokines and chemokines in the peripheral blood and used ELISA to determine both the cerebral and blood levels of interleukin-6 (IL-6). We also performed the chromatin immunoprecipitation (ChIP) assay to evaluate the extent of nuclear factor kappa-B (NF-κB) binding to the DNA elements in the IL-6 promoter regions. Also, we adopted the Western blotting assay to measure the cerebral levels of heat shock protein (HSP) 70, synapsin I, and ß-actin. Finally, we performed both histoimmunological and behavioral assessment to measure brain injury and neurological deficits, respectively. RESULTS: We first demonstrated that TBI upregulated nine pro-inflammatory and/or neurodegenerative messenger RNAs (mRNAs) in the peripheral blood such as CXCL10, IL-18, IL-16, Cd-70, Mif, Ppbp, Ltd, Tnfrsf 11b, and Faslg. In addition to causing neurological injuries, TBI also upregulated the following 14 anti-inflammatory and/or neuroregenerative mRNAs in the peripheral blood such as Ccl19, Ccl3, Cxcl19, IL-10, IL-22, IL-6, Bmp6, Ccl22, IL-7, Bmp7, Ccl2, Ccl17, IL-1rn, and Gpi. Second, we observed that EP inhibited both neurological injuries and six pro-inflammatory and/or neurodegenerative genes (Cxcl10, IL-18, IL-16, Cd70, Mif, and Faslg) but potentiated four anti-inflammatory and/or neuroregenerative genes (Bmp6, IL-10, IL-22, and IL-6). Prior depletion of cerebral HSP70 with gene silence significantly reversed the beneficial effects of EP in reducing neurological injuries and altered gene profiles after a TBI. A positive Pearson correlation exists between IL-6 and HSP70 in the peripheral blood or in the cerebral levels. In addition, gene silence of cerebral HSP70 significantly reduced the overexpression of NF-κB, IL-6, and synapsin I in the ipsilateral brain regions after an EP in rats. CONCLUSIONS: TBI causes neurological deficits associated with stimulating several pro-inflammatory gene profiles but inhibiting several anti-inflammatory gene profiles of cytokines and chemokines. Exercise protects against neurological injuries via stimulating an anti-inflammatory HSP70/NF-κB/IL-6/synapsin I axis in the injured brains.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Condicionamento Físico Animal/fisiologia , Sinapsinas/metabolismo , Animais , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/prevenção & controle , Masculino , Condicionamento Físico Animal/métodos , Distribuição Aleatória , Ratos , Ratos Wistar
10.
Chem Biol Interact ; 265: 47-54, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28104348

RESUMO

Heat stroke is characterized by hyperthermia, systemic inflammation, and multiple organ failure including arterial hypotension. This definition can be fulfilled by a rat model of heat stroke used in the present study. Anesthetized animals were exposed to heat exposure (43 °C for 70 min) and then returned to room temperature (26 °C) for recovery. One hour before heat exposure, an intraperitoneal dose of quercetin (30 mg/kg) or vehicle (normal saline 1 ml/kg) was administered to the experimental groups of rats. Additional injection was administered immediately after the onset of heat stroke. Immediately after the onset of heat stroke. Vehicle-treated rats displayed (i) hyperthermia; (ii) suppressed left ventricular function; (iii) decreased contents of cardiac total antioxiant capacity (e.g., superoxide dismutase, glutathione peroxidase, catalase); (iv) increased contents of cardiac oxidative capacity malondialdehyde and thiobarbituric acid reactive substances; (v) increased cardiac levels of pro-inflammatory cytokines tumor necrosis factor-α and interleukin-6; and (vi) decreased cardiac levels of an anti-inflammatory cytokine interleukin 10. Histopathologic and survival observation provided supportive evidence for biochemical analyses. These heat stroke reactions all can be significantly attenuated by quercetin therapy. Our data suggest that quercetin therapy might improve outcomes of heat stroke in rats by attenuating excessive hyperthermia as well as myocardial injury. The protective effects of quercetin could be attributed to anti-lipid peroxidative, anti-oxidant, and anti-inflammatory properties.


Assuntos
Cardiomiopatias/prevenção & controle , Golpe de Calor/complicações , Quercetina/farmacologia , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Citocinas/metabolismo , Hemodinâmica , Mediadores da Inflamação/metabolismo , Masculino , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley
11.
Cell Transplant ; 26(11): 1798-1810, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29338384

RESUMO

Microglia are the first source of a neuroinflammatory cascade, which seems to be involved in every phase of stroke-related neuronal damage. Two weeks after transient middle cerebral artery occlusion (MCAO), vehicle-treated rats displayed higher numbers of total ionized calcium-binding adaptor molecule 1 (Iba-1)-positive cells, greater cell body areas of Iba-1-positive cells, and higher numbers of hypertrophic Iba-1-positive cells (with a cell body area over 80 µm2) in the ipsilateral ischemic brain regions including the frontal cortex, striatum, and parietal cortex. In addition, MCAO decreased the number of migrating neuroblasts (or DCX- and 5-ethynyl-2'-deoxyuridine-positive cells) in the cortex, subventricular zone, and hippocampus of the ischemic brain, followed by neurological injury (including brain infarct and neurological deficits). Intravenous administration of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs; 1 × 106 or 4 × 106) at 24 h after MCAO reduced neurological injury, decreased the number of hypertrophic microglia/macrophages, and increased the number of newborn neurons in rat brains. Thus, the accumulation of hypertrophic microglia/macrophages seems to be detrimental to neurogenesis after stroke. Treatment with hUC-MSCs preserved adult newborn neurons and reduced functional impairment after transient cerebral ischemia by reducing the number of hypertrophic microglia/macrophages.


Assuntos
Isquemia Encefálica/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Mesenquimais/citologia , Neurônios/citologia , Cordão Umbilical/citologia , Análise de Variância , Animais , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Proteína Duplacortina , Humanos , Imuno-Histoquímica , Macrófagos/citologia , Macrófagos/fisiologia , Masculino , Células-Tronco Mesenquimais/fisiologia , Microglia/citologia , Microglia/fisiologia , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley
12.
Brain Behav ; 6(10): e00526, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27781140

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC) transplantation has been reported to improve neurological function following neural injury. Many physiological and molecular mechanisms involving MSC therapy-related neuroprotection have been identified. METHODS: A review is presented of articles that pertain to MSC therapy and diverse brain injuries including stroke, neural trauma, and heat stroke, which were identified using an electronic search (e.g., PubMed), emphasize mechanisms of MSC therapy-related neuroprotection. We aim to discuss neuroprotective mechanisms that underlie the beneficial effects of MSCs in treating stroke, neural trauma, and heatstroke. RESULTS: MSC therapy is promising as a means of augmenting brain repair. Cell incorporation into the injured tissue is not a prerequisite for the beneficial effects exerted by MSCs. Paracrine signaling is believed to be the most important mediator of MSC therapy in brain injury. The multiple mechanisms of action of MSCs include enhanced angiogenesis and neurogenesis, immunomodulation, and anti-inflammatory effects. Microglia are the first source of the inflammatory cascade during brain injury. Cytokines, including tumor necrosis factor-α, interleukin-1ß, and interleukin-6, are significantly produced by microglia in the brain after experimental brain injury. The proinflammatory M1 phenotype of microglia is associated with tissue destruction, whereas the anti-inflammatory M2 phenotype of microglia facilitates repair and regeneration. MSC therapy may improve outcomes of ischemic stroke, neural trauma, and heatstroke by inhibiting the activity of M1 phenotype of microglia but augmenting the activity of M2 phenotype of microglia. CONCLUSION: This review offers a testable platform for targeting microglial-mediated cytokines in clinical trials based upon the rational design of MSC therapy in the future. MSCs that are derived from the placenta provide a great choice for stem cell therapy. Although targeting the microglial activation is an important approach to reduce the burden of the injury, it is not the only one. This review focuses on this specific aspect.


Assuntos
Lesões Encefálicas/terapia , Golpe de Calor/terapia , Transplante de Células-Tronco Mesenquimais , Neuroproteção/fisiologia , Acidente Vascular Cerebral/terapia , Animais , Lesões Encefálicas/fisiopatologia , Golpe de Calor/fisiopatologia , Humanos , Células-Tronco Mesenquimais/fisiologia , Acidente Vascular Cerebral/fisiopatologia
13.
Mol Neurobiol ; 53(10): 7329-7340, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26696494

RESUMO

ß-amyloid (Aß)-mediated neuronal apoptosis contributes to the pathogenesis of Alzheimer's disease (AD). This study aimed to investigate whether astragalosides (AST) could inhibit Aß-induced apoptosis in vivo and in vitro and to explore the underlying mechanisms. Amyloid ß-protein fragment 25-35 (Aß25-35) was administered to cerebral lateral ventricle of rats to make the AD models in vivo. AST was able to attenuate both cortical cell degeneration and memory deficits in the AD rats. AST also inhibited Aß25-35-induced cytotoxicity (e.g., decreased cell viability); apoptosis (e.g., increased caspase-3 expression, increased DNA fragmentation, and Tau hyperphosphorylation); synaptotoxicity (e.g., increased loss of both a dendritic marker, microtubule-associated protein 2 (MAP-2) and synaptic proteins, synaptophysins); and mitochondrial dysfunction (e.g., increased mitochondrial membrane potential) in cultured primary rat cortical cells. The beneficial effect of AST in reducing Aß-induced cytotoxicity, apoptosis, and mitochondrial dysfunction in cortical cells were blocked by inhibition of phosphoinositide 3-kinase (PI3K)-dependent protein kinase B (PKB, as known as AKT) activation with LY294002. In addition, inhibition of extracellular protein kinase (ERK) with U0126 shared with the AST the same beneficial effects in reducing Aß-induced apoptosis. Our data suggest that the cortical PI3K/AKT and MAPK (or ERK) pathways as appealing therapeutic targets in treating AD, and AST may have a positive impact on AD treatment via modulation of both PI3K/AKT and ERK pathways.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Córtex Cerebral/patologia , Saponinas/uso terapêutico , Doença de Alzheimer/complicações , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Caspase 3/metabolismo , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Masculino , Transtornos da Memória/complicações , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Degeneração Neural/tratamento farmacológico , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Saponinas/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Proteínas tau/metabolismo
14.
Mol Neurobiol ; 53(7): 4961-71, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26374550

RESUMO

Ischemic stroke, caused by obstruction of blood flow to the brain, would initiate microglia activation which contributes to neuronal damage. Therefore, inhibition of microglia-mediated neuroinflammation could be a therapeutic strategy for ischemic stroke. This study was aimed to elucidate the anti-inflammatory effects of alpha-lipoic acid and etanercept given either singly or in combination in rats subjected to middle cerebral artery occlusion. Both α-lipoic acid and etanercept markedly reduced cerebral infarct, blood-brain barrier disruption, and neurological motor deficits with the former drug being more effective with the dosage used. Furthermore, when used in combination, the reduction was more substantial. Remarkably, a greater diminution in the serum levels of tumor necrosis factor-alpha as well as the brain levels of microglial activation (e.g., microgliosis, amoeboid microglia, and microglial overexpression of tumor necrosis factor-α) was observed with the combined drug treatment as compared to the drugs given separately. We conclude that inhibition of peripheral tumor necrosis factor-alpha as well as downregulation of brain microglial activation by alpha-lipoic acid or etanercept protect rat brain against ischemic stroke. Moreover, when both drugs were used in combination, the stroke recovery was promoted more extensively.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Etanercepte/administração & dosagem , Acidente Vascular Cerebral/metabolismo , Ácido Tióctico/administração & dosagem , Fator de Necrose Tumoral alfa/metabolismo , Animais , Antioxidantes/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Isquemia Encefálica/patologia , Isquemia Encefálica/prevenção & controle , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Quimioterapia Combinada , Masculino , Fármacos Neuroprotetores/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/prevenção & controle , Fator de Necrose Tumoral alfa/antagonistas & inibidores
15.
J Formos Med Assoc ; 114(4): 328-38, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25839766

RESUMO

BACKGROUND/PURPOSE: The primary goal of this study was to test whether high-altitude exposure (HAE: 0.9% O(2) at 0.47 ATA for 24 hours) was capable of increasing the systemic inflammatory markers as well as the toxic organ injury indicators in rats, with a secondary goal to test whether preinduction of heat shock protein (HSP) 70 by hypobaric hypoxia preconditioning (HHP: 18.3% O(2) at 0.66 ATA for 5 h/day on 5 days consecutively for 2 weeks) attenuated the proposed increased serum levels of both the systemic inflammatory markers and the toxic organ injury indicators. METHODS: Rats were assigned to: (1) non-HHP (21% O(2) at 1.0 ATA)+non-HAE (21% O(2) at 1.0 ATA) group; (2) non-HHP+HAE group; (3) HHP+non-HAE group; (4) HHP+HAE group; and (5) HHP+HSP70 antibodies (Ab)+HAE group. For the HSP70Ab group, a neutralizing HSP70Ab was injected intravenously at 24 hours prior to HAE. All the physiological and biochemical parameters were obtained at the end of HAE or the equivalent time period of non-HAE. Blood samples were obtained for determination of both the systemic inflammatory markers (e.g., serum tumor necrosis factor-α, interleukin-1ß, E-selectin, intercellular adhesion molecule-1, and liver myeloperoxidase activity) and the toxic organ injury indicators (e.g., nitric oxide metabolites, 2,3-dihydroxybenzoic acid, and lactate dehydrogenase). RESULTS: HHP, in addition to inducing overexpression of tissue HSP70, significantly attenuated the HAE-induced hypotension, bradycardia, hypoxia, acidosis, and increased tissue levels of both the systemic inflammatory markers and the toxic organ injury indicators. The beneficial effects of HHP in inducing tissue overexpression of HSP70 as well as in preventing the HAE-induced increased levels of the systemic inflammatory markers and the toxic organ injury indicators could be significantly reduced by HSP70Ab preconditioning. CONCLUSION: These results suggest that HHP may downgrade both the systemic inflammatory markers and the toxic organ injury indicators in HAE by upregulating tissue HSP70.


Assuntos
Doença da Altitude/sangue , Biomarcadores/sangue , Proteínas de Choque Térmico HSP70/administração & dosagem , Animais , Modelos Animais de Doenças , Selectina E/sangue , Hidroxibenzoatos/sangue , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/sangue
16.
Cell Transplant ; 24(5): 921-37, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24612725

RESUMO

Stem cells from human exfoliated deciduous tooth pulp (SHED) is a promising approach for the treatment of stroke and spinal cord injury. In this study, we investigated the therapeutic effects of SHED for the treatment of multiple organ (including brain, particularly hypothalamus) injury in heatstroke mice. ICR male mice were exposed to whole body heating (WBH; 41.2°C, relative humidity 50-55%, for 1 h) and then returned to normal room temperature (26°C). We observed that intravenous administration of SHED immediately post-WBH exhibited the following therapeutic benefits for recovery after heatstroke: (a) inhibition of WBH-induced neurologic and thermoregulatory deficits; (b) reduction of WBH-induced ischemia, hypoxia, and oxidative damage to the brain (particularly the hypothalamus); (c) attenuation of WBH-induced increased plasma levels of systemic inflammatory response molecules, such as tumor necrosis factor-α and intercellular adhesion molecule-1; (d) improvement of WBH-induced hypothalamo-pituitary-adrenocortical (HPA) axis activity (as reflected by enhanced plasma levels of both adrenocorticotrophic hormone and corticosterone); and (e) attenuation of WBH-induced multiple organ apoptosis as well as lethality. In conclusion, post-WBH treatment with SHED reduced induction of proinflammatory cytokines and oxidative radicals, enhanced plasma induction of both adrenocorticotrophic hormone and corticosterone, and improved lethality in mouse heatstroke. The protective effect of SHED may be related to a decreased inflammatory response, decreased oxidative stress, and an increased HPA axis activity following the WBH injury.


Assuntos
Polpa Dentária/metabolismo , Golpe de Calor/terapia , Transplante de Células-Tronco , Células-Tronco/metabolismo , Animais , Criança , Feminino , Golpe de Calor/sangue , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR
17.
Curr Med Chem ; 22(6): 759-70, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25386818

RESUMO

Microglia and several inflammatory cytokines and neurotrophic growth factors are involved in traumatic brain injury (TBI). Tumor necrosis factor-alpha (TNF-α) can be released by microglia, astrocytes, and neurons. TNF-α has been reported to be both proneurogenic and antineurogenic, depending upon the model, method, and cell-derived region. There are two subtypes of microglia: M1 and M2. The former (or M1 subtype of non-phagocytic microglia) is able to secrete higher levels of TNF-α but lower levels of interleukin (IL)-10 (IL-10), an anti-inflammatory cytokine. Both the proinflammatory and the pro-apoptotic function can also be promoted by activation of tumor necrosis factor-receptor 1 (TNF-R1). In contrast, M2 activation produces lower levels of TNF-α but higher levels of IL-10. Pro-growth and survival pathways can be promoted by the activation of TNFR2. During the acute stage of TBI, both M1 subtype of microglia and TNF-R1 are activated to cause higher levels of TNF-α but lower levels of IL-10, which lead to suppressed neurogenesis, neuronal loss and organ dysfunction (so-called microglial activation I). In contrast, activation of both M2 subtype of microglia and TNF-R2 is able to promote neurogenesis and tissue recovery (so-called microglial activation II). The severity of TBI depends upon the net effects between microglial activation I and microglial activation II. Indeed, by using rodent models of TBI, therapeutic evaluation studies reveal that several agents or strategies attenuate contused brain volume and neurological deficits by inhibiting microglial activation I but inducing microglial activation II. For example, etanercept therapy might attenuate contused brain volume and neurological deficits by inactivating the M1 subtype and TNF-R1 to reduce the microglial activation I response, but it might promote neurogenesis and functional recovery by activating the M2 subtype and TNF-R2. Therefore, based on microglial responses I and II, we conclude that future studies should focus on multiple therapeutic agents and strategies for optimal TBI therapy.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Microglia/efeitos dos fármacos , Microglia/patologia , Terapia de Alvo Molecular/métodos , Doença Aguda , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Humanos , Microglia/metabolismo
18.
J Neuroimmune Pharmacol ; 10(1): 45-54, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25331680

RESUMO

Traumatic brain injury (TBI) causes increased release of several mediators from injured and dead cells and elicits microglial activation. Activated microglia change their morphology, migrate to injury sites, and release tumor necrosis factor-alpha (TNF-α) and others. In this study we used a controlled fluid percussion injury model of TBI in the rat to determine whether early (4 h post-injury) or late (4 days post-injury) treatment with MLC 601, a Traditional Chinese Medicine, would affect microglial activation and improve recovery. MLC 601 was chosen for this study because its herbal component MLC 901 was beneficial in treating TBI in rats. Herein, rats with induced TBI were treated with MLC 601 (0.2-0.8 mg/kg) 1 h (early treatment) or 4 day post-injury (late treatment) and then injected once daily for consecutive 2 days. Acute neurological and motor deficits were assessed in all rats the day before and 4 days after early MLC 601 treatment. An immunofluorescence microscopy method was used to count the numbers of the cells colocalized with neuron- and apoptosis-specific markers, and the cells colocalized with microglia- and TNF-α-specific markers, in the contused brain regions 4 days post-injury. An immunohistochemistry method was used to evaluate both the number and the morphological transformation of microglia in the injured areas. It was found that early treatment with MLC 601 had better effects in reducing TBI-induced cerebral contusion than did the late therapy with MLC 601. Cerebral contusion caused by TBI was associated with neurological motor deficits, brain apoptosis, and activated microglia (e.g., microgliosis, amoeboid microglia, and microglial overexpression of TNF-α), which all were significantly attenuated by MLC 601 therapy. Our data suggest that MLC 601 is a promising agent for treatment of TBI in rats.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Medicamentos de Ervas Chinesas/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Contagem de Células , Transtornos Neurológicos da Marcha/etiologia , Transtornos Neurológicos da Marcha/prevenção & controle , Imuno-Histoquímica , Masculino , Microglia/efeitos dos fármacos , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/prevenção & controle , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos
19.
J Trauma Acute Care Surg ; 77(4): 585-91, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25250598

RESUMO

BACKGROUND: The primary goal of this study was to test whether high-altitude exposure (HAE of 9.7% O2 at 0.47 absolute atmosphere [ATA] for 3 days) was capable of increasing lung edema, neutrophil, and hemorrhage scores as well as decreasing lung levels of both aquaporin 1 (AQP1) and AQP5 proteins and messenger RNA (mRNA) expression in rats, with a secondary goal to test whether a preinduction of heat shock protein 70 (HSP70) by hyperbaric oxygen preconditioning (HBO2P of 100% O2 at 2.0 ATA for 1 hour per day for 5 consecutive days) attenuated the HAE-induced increased lung injury scores and decreased lung AQP1 and AQP5 protein and mRNA expressions. METHODS: Rats were assigned to (1) non-HBO2P (21% O2 at 1.0 ATA) + non-HAE (21% O2 at 1.0 ATA) group; (2) non-HBO2P + HAE group; (3) HBO2P + HAE group; and HBO2P + HSP70 antibodies (Ab) + HAE group. For the HSP70 Ab group, a neutralizing HSP70 Ab was injected intravenously at 24 hours before HAE. All the physiologic and biochemical parameters were obtained at the end of HAE or the equivalent period of non-HAE. The cardiovascular and blood gas parameters were monitored for all experiments. Bronchoalveolar lavage (BAL) was performed to determine proinflammatory cytokines (interleukin 6, interleukin 1ß, and tumor necrosis factor α). Parts of the lung were excised for myeloperoxidase activity measurement, whereas the rest was collected for lung damage score assessments. AQP1 and AQP5 protein and mRAN expressions were also determined in the lung tissues. RESULTS: In the non-HBO2P + HAE group, the animals displayed higher values of lung myeloperoxidase activity, BAL proinflammatory cytokines, lung water weight, and acute lung injury scores compared with those of the non-HBO2P + non-HAE controls. In contrast, the non-HBO2P + HAE group rats had lower values of lung AQP1 and AQP5 protein and mRNA expressions, mean arterial pressure, heart rate, SO2, Paco2, HCO3, and pH compared with those of non-HBO2P + non-HAE group rats. The increased acute lung edema, neutrophil, and hemorrhage scores; increased BAL levels of proinflammatory cytokines; decreased lung AQP1 and AQP5 protein and mRNA expressions; and hypotension, bradycardia, hypoxia, and acidosis caused by HAE were all significantly attenuated by HBO2P. CONCLUSION: Our data indicate that HBO2P may attenuate high-altitude acute lung injury by a preinduction of lung HSP70 in rats.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Altitude , Proteínas de Choque Térmico HSP70/metabolismo , Precondicionamento Isquêmico/métodos , Edema Pulmonar/prevenção & controle , Animais , Aquaporina 1/metabolismo , Aquaporina 5/metabolismo , Western Blotting , Oxigenoterapia Hiperbárica , Masculino , Edema Pulmonar/metabolismo , Ratos , Ratos Wistar
20.
Cell Mol Neurobiol ; 34(6): 825-37, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24807460

RESUMO

Astragalosides (AST) are reported to be neuroprotective in focal cerebral ischemic models in vivo. In this study, the direct effect of AST against oxygen and glucose deprivation (OGD) including neuronal injury and the underlying mechanisms in vitro were investigated. 5 h OGD followed by 24 h of reperfusion [adding back oxygen and glucose (OGD-R)] was used to induce in vitro ischemia reperfusion injury in differentiated rat pheochromocytoma PC12 cells. AST (1, 100, and 200 µg/mL) were added to the culture after 5 h of the OGD ischemic insult and was present during the reoxygenation phases. A key finding was that OGD-R decreased cell viability, increased lactate dehydrogenase, increased reactive oxygen species, apoptosis, autophagy, functional impairment of mitochondria, and endoplasmic reticulum stress in PC12 cells, all of which AST treatment significantly reduced. In addition, AST attenuated OGD-R-induced cell loss through P38 MAPK activation a neuroprotective effect blunted by SB203580, a specific inhibitor of P38 MAPK. Our data suggest that both apoptosis and autophagy are important characteristics of OGD-R-induced PC12 death and that treating PC12 cells with AST blocked OGD-R-induced apoptosis and autophagy by suppressing intracellular oxidative stress, functional impairment of mitochondria, and endoplasmic reticulum stress. Our data provide identification of AST that can concomitantly inhibit multiple cells death pathways following OGD injuries in neural cells.


Assuntos
Glucose/metabolismo , Fármacos Neuroprotetores/farmacologia , Oxigênio/metabolismo , Saponinas/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Triterpenos/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Mitocôndrias/metabolismo , Células PC12 , Ratos , Traumatismo por Reperfusão , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA