Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Int J Mol Sci ; 22(2)2021 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-33430045

RESUMO

Plant-based nutritional supplementation has been shown to attenuate and reduce mortality in the processes of both acute and chronic disorders, including diabetes, obesity, cardiovascular disease, cancer, inflammatory diseases, and neurological and neurodegenerative disorders. Low-level systemic inflammation is an important contributor to these afflictions and diets enriched in phytochemicals can slow the progression. The goal of this study was to determine the impact of lipopolysaccharide (LPS)-induced inflammation on changes in glucose and insulin tolerance, performance enhancement, levels of urinary neopterin and concentrations of neurotransmitters in the striatum in mouse models. Both acute and chronic injections of LPS (2 mg/kg or 0.33 mg/kg/day, respectively) reduced glucose and insulin tolerance and elevated neopterin levels, which are indicative of systemic inflammatory responses. In addition, there were significant decreases in striatal neurotransmitter levels (dopamine and DOPAC), while serotonin (5-HT) levels were essentially unchanged. LPS resulted in impaired execution in the incremental loading test, which was reversed in mice on a supplemental plant-based diet, improving their immune function and maintaining skeletal muscle mitochondrial activity. In conclusion, plant-based nutritional supplementation attenuated the metabolic changes elicited by LPS injections, causing systemic inflammatory activity that contributed to both systemic and neurological alterations.


Assuntos
Inflamação/dietoterapia , Músculo Esquelético/metabolismo , Obesidade/dietoterapia , Compostos Fitoquímicos/farmacologia , Animais , Dieta , Suplementos Nutricionais , Modelos Animais de Doenças , Dopamina/metabolismo , Glucose/metabolismo , Inflamação/induzido quimicamente , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Camundongos , Músculo Esquelético/efeitos dos fármacos , Neopterina/urina , Obesidade/induzido quimicamente , Obesidade/patologia , Serotonina/metabolismo
2.
J Neurosci ; 39(47): 9465-9476, 2019 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-31611307

RESUMO

Serum amyloid A (SAA) proteins are acute-phase reactant associated with high-density lipoprotein (HDL) particles and increase in the plasma 1000-fold during inflammation. Recent studies have implicated SAAs in innate immunity and various disorders; however, the precise mechanism eludes us. Previous studies have shown SAAs are elevated following stroke and cerebral ischemia, and our studies demonstrated that SAA-deficient mice reduce inflammation and infarct volumes in a mouse stroke model. Our studies demonstrate that SAA increases the cytokine interleukin-1ß (IL-1ß), which is mediated by Nod-like receptor protein 3 (NLRP3) inflammasome, cathepsin B, and caspase-1 and may play a role in the pathogenesis of neurological disorders. SAA induced the expression of NLRP3, which mediated IL-1ß induction in murine BV-2 cells and both sex primary mouse microglial cells, in a dose- and time-dependent fashion. Inhibition or KO of the NLRP3 in microglia prevented the increase in IL-1ß. N-acetyl-l-cysteine and mito-TEMPO blocked the induction of IL-1ß by inhibiting ROS with SAA treatment. In addition, inhibition of cathepsin B with different drugs or microglia from CatB-deficient mice attenuated inflammasome activation. Our studies suggest that the impact of SAA on inflammasome stimulation is mediated in part by the receptor for advanced glycation endproducts and Toll-like receptor proteins 2 and 4. SAA induced inflammatory cytokines and an M1 phenotype in the microglial cells while downregulating anti-inflammation M2 phenotype. These studies suggest that brain injury to can elicit a systemic inflammatory response mediated through SAA that contributes to the pathological outcomes.SIGNIFICANCE STATEMENT In the present study, serum amyloid A can induce that activation of the inflammasome in microglial cells and give rise to IL-1ß release, which can further inflammation in the brain following neurological diseases. The also presents a novel target for therapeutic approaches in stroke.


Assuntos
Isquemia Encefálica/metabolismo , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteína Amiloide A Sérica/toxicidade , Animais , Isquemia Encefálica/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia
3.
Biochem Biophys Res Commun ; 523(3): 678-684, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-31948754

RESUMO

The promotion of axonal regeneration is required for functional recovery from stroke and various neuronal injuries. However, axonal regeneration is inhibited by diverse axonal growth inhibitors, such as Nogo-A. Nogo-66, a C-terminal domain of Nogo-A, binds to the Nogo-A receptor 1 (NgR1) and induces the collapse of growth cones and inhibits neurite outgrowth. NgR1 is also a receptor for additional axonal growth inhibitors, suggesting it is an important target for the prevention of axonal growth inhibition. By using the indirect immunofluorescence method, we show for the first time that a cell-permeable cAMP analog (dibutyryl-cAMP) induced a rapid decrease in the cell surface expression of NgR1 in Neuroscreen-1 (NS-1) cells. The biotinylation method revealed that cAMP indeed induced internalization of NgR1 within minutes. Other intracellular cAMP-elevating agents, such as forskolin, which directly activates adenylyl cyclase, and rolipram, which inhibits cyclic nucleotide phosphodiesterase, also induced this process. This internalization was found to be reversible and influenced by intracellular levels of cAMP. Using selective activators and inhibitors of protein kinase A (PKA) and the exchange protein directly activated by cAMP (Epac), we found that NgR1 internalization is independent of PKA, but dependent on Epac. The decrease in cell surface expression of NgR1 desensitized NS-1 cells to Nogo-66-induced growth cone collapse. Therefore, it is likely that besides axonal growth inhibitors affecting neurons, neurons themselves also self-regulate their sensitivity to axonal growth inhibitors, as influenced by intracellular cAMP/Epac. This normal cellular regulatory mechanism may be pharmacologically exploited to overcome axonal growth inhibitors, and enhance functional recovery after stroke and neuronal injuries.


Assuntos
AMP Cíclico/metabolismo , Cones de Crescimento/metabolismo , Neurônios/metabolismo , Proteínas Nogo/metabolismo , Receptor Nogo 1/metabolismo , Animais , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neurônios/citologia , Células PC12 , Transporte Proteico , Ratos
4.
Clin Exp Hypertens ; 42(7): 622-639, 2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-32420765

RESUMO

PURPOSE: To review the recent developments on the effect of chronic high mean arterial blood pressure (MAP) on cerebral blood flow (CBF) autoregulation and supporting the notion that CBF autoregulation impairment has connection with chronic cerebral diseases. Method: A narrative review of all the relevant papers known to the authors was conducted. Results: Our understanding of the connection between cerebral perfusion impairment and chronic high MAP and cerebral disease is rapidly evolving, from cerebral perfusion impairment being the result of cerebral diseases to being the cause of cerebral diseases. We now better understand the intertwined impact of hypertension and Alzheimer's disease (AD) on cerebrovascular sensory elements and recognize cerebrovascular elements that are more vulnerable to these diseases. Conclusion: We conclude with the suggestion that the sensory elements pathology plays important roles in intertwined mechanisms of chronic high MAP and AD that impact cerebral perfusion.


Assuntos
Doença de Alzheimer/fisiopatologia , Pressão Arterial/fisiologia , Circulação Cerebrovascular , Hipertensão/fisiopatologia , Homeostase , Humanos
5.
Pediatr Res ; 86(5): 662-669, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30712059

RESUMO

BACKGROUND: Polymorphic alleles of the vitamin D (vitD)-binding protein (VDBP) gene are associated with discriminatory differences in circulating concentrations of 25-hydroxyvitamin D (25-D), the indicator of vitD status (sufficiency defined by the Endocrine Society as ≥75 nmol/L). Within a diverse group of children, we hypothesized that reaching recommended daily allowance (RDA) of vitD intake would have differential impact on vitD status depending on VDBP variability. METHODS: VDBP alleles (Gc1S, Gc1F, Gc2) in 123 children (1-4 annual visits/child; ages 1-8 years) were compared for relationships with serum 25-D concentrations and daily vitD intake. RESULTS: In African-American children, reaching the vitD RDA was associated with significantly higher mean serum 25-D concentrations for the 20% carrying the VDBP 1S allele than for the large majority without this allele (77 vs. 61 nmol/L 25-D; p = 0.038). Children with the Gc1S/1S homozygous genotype (30% Caucasians, 24% Hispanics, 2% African-Americans) who met RDA had 51% (39 nmol/L) greater mean serum 25-D than those below RDA (p < 0.0001). CONCLUSIONS: VDBP genetic variability was a significant factor affecting childhood vitD status when following RDA guidelines. This study may inform public health policy of uniformity in recommended childhood vitD dosage, especially regarding racially/ethnically associated disparities.


Assuntos
Política Nutricional , Proteína de Ligação a Vitamina D/sangue , Vitamina D/administração & dosagem , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Vitamina D/sangue
6.
J Neuroinflammation ; 15(1): 277, 2018 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-30249250

RESUMO

BACKGROUND: Repetitive mild traumatic brain injuries (rmTBI) are associated with cognitive deficits, inflammation, and stress-related events. We tested the effect of nutrient intake on the impact of rmTBI in an animal model of chronic traumatic encephalopathy (CTE) to study the pathophysiological mechanisms underlying this model. We used a between group design rmTBI closed head injuries in mice, compared to a control and nutrient-treated groups. METHODS: Our model allows for controlled, repetitive closed head impacts to mice. Briefly, 24-week-old mice were divided into five groups: control, rmTBI, and rmTBI with nutrients (2% of NF-216, NF-316 and NF-416). rmTBI mice received four concussive impacts over 7 days. Mice were treated with NutriFusion diets for 2 months prior to the rmTBI and until euthanasia (6 months). Mice were then subsequently euthanized for macro- and micro-histopathologic analysis for various times up to 6 months after the last TBI received. Animals were examined behaviorally, and brain sections were immunostained for glial fibrillary acidic protein (GFAP) for astrocytes, iba-1 for activated microglia, and AT8 for phosphorylated tau protein. RESULTS: Animals on nutrient diets showed attenuated behavioral changes. The brains from all mice lacked macroscopic tissue damage at all time points. The rmTBI resulted in a marked neuroinflammatory response, with persistent and widespread astrogliosis and microglial activation, as well as significantly elevated phospho-tau immunoreactivity to 6 months. Mice treated with diets had significantly reduced inflammation and phospho-tau staining. CONCLUSIONS: The neuropathological findings in the rmTBI mice showed histopathological hallmarks of CTE, including increased astrogliosis, microglial activation, and hyperphosphorylated tau protein accumulation, while mice treated with diets had attenuated disease process. These studies demonstrate that consumption of nutrient-rich diets reduced disease progression.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Inflamação/etiologia , Inflamação/terapia , Nutrientes/uso terapêutico , Tauopatias/etiologia , Tauopatias/terapia , Animais , Sintomas Comportamentais/etiologia , Sintomas Comportamentais/terapia , Lesões Encefálicas Traumáticas/terapia , Citocinas/metabolismo , Modelos Animais de Doenças , Elevação dos Membros Posteriores/fisiologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Força Muscular/fisiologia , Assunção de Riscos , Sono/fisiologia , Aprendizagem Espacial/fisiologia , Natação/psicologia , Índices de Gravidade do Trauma , Proteínas tau/genética
7.
Ethn Dis ; 28(1): 61-68, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29467568

RESUMO

Racial-ethnic disparities in stroke recovery are well-established in the United States but the underlying causes are not well-understood. The typical assumption that racial-ethnic disparities in stroke recovery are explained by health care access inequities may be simplistic as access to stroke-related rehabilitation, for example, does not adequately explain the observed disparities. To approach the problem in a more comprehensive fashion, the Wide Spectrum Investigation of Stroke Outcome Disparities on Multiple Levels (WISSDOM) was developed to bring together scientists from Regenerative Medicine, Neurology, Rehabilitation, and Nursing to examine disparities in stroke "recovery." As a result, three related projects (basic science, clinical science and population science) were designed utilizing animal modeling, mapping of brain connections, and community-based interventions. In this article we describe: 1) the goals and objectives of the individual projects; and 2) how these projects could provide critical evidence to explain why racial-ethnic minorities traditionally experience recovery trajectories that are worse than Whites.


Assuntos
Etnicidade , Disparidades nos Níveis de Saúde , Grupos Raciais , Reabilitação do Acidente Vascular Cerebral , Acidente Vascular Cerebral/etnologia , Pesquisa Biomédica , Objetivos , Humanos , Avaliação de Resultados em Cuidados de Saúde , Recuperação de Função Fisiológica , Reabilitação do Acidente Vascular Cerebral/métodos , Estados Unidos
8.
J Biol Chem ; 291(4): 1957-1973, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26620563

RESUMO

Experimental evidence supports the role of mitochondrial ceramide accumulation as a cause of mitochondrial dysfunction and brain injury after stroke. Herein, we report that SIRT3 regulates mitochondrial ceramide biosynthesis via deacetylation of ceramide synthase (CerS) 1, 2, and 6. Reciprocal immunoprecipitation experiments revealed that CerS1, CerS2, and CerS6, but not CerS4, are associated with SIRT3 in cerebral mitochondria. Furthermore, CerS1, -2, and -6 are hyperacetylated in the mitochondria of SIRT3-null mice, and SIRT3 directly deacetylates the ceramide synthases in a NAD(+)-dependent manner that increases enzyme activity. Investigation of the SIRT3 role in mitochondrial response to brain ischemia/reperfusion (IR) showed that SIRT3-mediated deacetylation of ceramide synthases increased enzyme activity and ceramide accumulation after IR. Functional studies demonstrated that absence of SIRT3 rescued the IR-induced blockade of the electron transport chain at the level of complex III, attenuated mitochondrial outer membrane permeabilization, and decreased reactive oxygen species generation and protein carbonyls in mitochondria. Importantly, Sirt3 gene ablation reduced the brain injury after IR. These data support the hypothesis that IR triggers SIRT3-dependent deacetylation of ceramide synthases and the elevation of ceramide, which could inhibit complex III, leading to increased reactive oxygen species generation and brain injury. The results of these studies highlight a novel mechanism of SIRT3 involvement in modulating mitochondrial ceramide biosynthesis and suggest an important role of SIRT3 in mitochondrial dysfunction and brain injury after experimental stroke.


Assuntos
Lesões Encefálicas/enzimologia , Proteínas de Membrana/metabolismo , Mitocôndrias/enzimologia , Sirtuína 3/metabolismo , Esfingosina N-Aciltransferase/metabolismo , Animais , Apoptose , Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Ceramidas/metabolismo , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Estresse Oxidativo , Sirtuína 3/genética , Esfingosina N-Aciltransferase/genética
9.
J Transl Med ; 14: 1, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26727970

RESUMO

BACKGROUND: Glioblastoma (GBM) is the deadliest of brain tumors. Standard treatment for GBM is surgery, followed by combined radiation therapy and chemotherapy. Current therapy prolongs survival but does not offer a cure. We report on a novel immunotherapy against GBM, tested in an animal model of C57BL/6 mice injected intra-cranially with a lethal dose of murine GL261 glioma cells. METHODS: Ten week-old C57BL/6 mice were anesthetized before injection of 2 × 10(4) GL261 cells in the right cerebral hemisphere and after 3 days half of the mice were administered a single subcutaneous (s.c.) injection of irradiated semi-allogeneic vaccine, while mock-vaccinated mice received a s.c. injection of phosphate-buffered saline (PBS). Tumor engraftment was monitored through bioluminescence imaging (BLI). Length of animal survival was measured by Kaplan-Meier graphs and statistics. At time of sacrifice brain tissue was processed for estimation of tumor size and immunohistochemical studies. RESULTS: Overall survival of vaccinated mice was significantly longer compared to mock-vaccinated mice. Five to ten percent of vaccinated mice survived more than 90 days following the engraftment of GL261 cells in the brain and appeared to be free of disease by BLI. Tumor volume in the brain of vaccinated mice was on average five to ten-fold smaller compared to mock-vaccinated mice. In vaccinated mice, conspicuous microglia infiltrates were observed in tumor tissue sections and activated microglia appeared to form a fence along the perimeter of the tumor cells. The results of these animal studies persuaded the Office of Orphan Products Development of the Food and Drug Administration (FDA) to grant Orphan Drug Designation for treatment of GBM with irradiated, semi-allogeneic vaccines. CONCLUSIONS: Our preclinical observations suggest that semi-allogeneic vaccines could be tested clinically on subjects with GBM, as an adjuvant to standard treatment.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/imunologia , Vacinas Anticâncer/uso terapêutico , Glioma/tratamento farmacológico , Glioma/imunologia , Animais , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Glioma/patologia , Estimativa de Kaplan-Meier , Medições Luminescentes , Masculino , Camundongos Endogâmicos C57BL , Peroxidase/metabolismo , Coloração e Rotulagem , Vacinação
10.
J Biol Chem ; 289(19): 13142-54, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24659784

RESUMO

In addition to immediate brain damage, traumatic brain injury (TBI) initiates a cascade of pathophysiological events producing secondary injury. The biochemical and cellular mechanisms that comprise secondary injury are not entirely understood. Herein, we report a substantial deregulation of cerebral sphingolipid metabolism in a mouse model of TBI. Sphingolipid profile analysis demonstrated increases in sphingomyelin species and sphingosine concurrently with up-regulation of intermediates of de novo sphingolipid biosynthesis in the brain. Investigation of intracellular sites of sphingosine accumulation revealed an elevation of sphingosine in mitochondria due to the activation of neutral ceramidase (NCDase) and the reduced activity of sphingosine kinase 2 (SphK2). The lack of change in gene expression suggested that post-translational mechanisms are responsible for the shift in the activities of both enzymes. Immunoprecipitation studies revealed that SphK2 is complexed with NCDase and cytochrome oxidase (COX) subunit 1 in mitochondria and that brain injury hindered SphK2 association with the complex. Functional studies showed that sphingosine accumulation resulted in a decreased activity of COX, a rate-limiting enzyme of the mitochondrial electron transport chain. Knocking down NCDase reduced sphingosine accumulation in mitochondria and preserved COX activity after the brain injury. Also, NCDase knockdown improved brain function recovery and lessened brain contusion volume after trauma. These studies highlight a novel mechanism of secondary TBI involving a disturbance of sphingolipid-metabolizing enzymes in mitochondria and suggest a critical role for mitochondrial sphingosine in promoting brain injury after trauma.


Assuntos
Ceramidase Alcalina/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Esfingosina/metabolismo , Ceramidase Alcalina/genética , Animais , Encéfalo/patologia , Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Proteínas do Tecido Nervoso/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/genética
11.
J Neuroinflammation ; 12: 247, 2015 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-26714866

RESUMO

BACKGROUND: Complement promotes neuroinflammation and injury in models of stroke. However, complement is also being increasingly implicated in repair and regeneration after central nervous system (CNS) injury, and some complement deficiencies have been shown to provide acute, but not subacute, protection after murine stroke. Here, we investigate the dual role of complement in injury and repair after cerebral ischemia and reperfusion. METHODS: We used complement-deficient mice and different complement inhibitors in a model of transient middle cerebral artery occlusion to investigate complement-dependent cellular and molecular changes that occur through the subacute phase after stroke. RESULTS: C3 deficiency and site-targeted complement inhibition with either CR2-Crry (inhibits all pathways) or CR2-fH (inhibits alternative pathway) significantly reduced infarct size, reduced apoptotic cell death, and improved neurological deficit score in the acute phase after stroke. However, only in CR2-fH-treated mice was there sustained protection with no evolution of injury in the subacute phase. Whereas both inhibitors significantly reduced microglia/macrophage activation and astrogliosis in the subacute phase, only CR2-fH improved neurological deficit and locomotor function, maintained neurogenesis markers, enhanced neuronal migration, and increased VEGF expression. These findings in CR2-fH-treated mice correlated with improved performance in spatial learning and passive avoidance tasks. The complement anaphylatoxins have been implicated in repair and regenerative mechanisms after CNS injury, and in this context CR2-fH significantly reduced, but did not eliminate the generation of C5a within the brain, unlike CR2-Crry that completely blocked C5a generation. Gene expression profiling revealed that CR2-fH treatment downregulated genes associated with apoptosis, TGFß signaling, and neutrophil activation, and decreased neutrophil infiltration was confirmed by immunohistochemistry. CR2-fH upregulated genes for neural growth factor and mediators of neurogenesis and neuronal migration. Live animal imaging demonstrated that following intravenous injection, CR2-fH targeted specifically to the post-ischemic brain, with a tissue half-life of 48.5 h. Finally, unlike C3 deficiency, targeted complement inhibition did not increase susceptibility to lethal post-stroke infection, an important consideration for stroke patients. CONCLUSIONS: Ischemic brain tissue-targeted and selective inhibition of alternative complement pathway provide self-limiting inhibition of complement activation and reduces acute injury while maintaining complement-dependent recovery mechanisms into the subacute phase after stroke.


Assuntos
Via Alternativa do Complemento/fisiologia , Marcação de Genes/métodos , Receptores de Complemento 3d/deficiência , Receptores de Complemento 3d/genética , Regeneração/fisiologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Acidente Vascular Cerebral/prevenção & controle
13.
J Immunol ; 188(3): 1460-8, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22198950

RESUMO

Self-reactive natural Abs initiate injury following ischemia and reperfusion of certain tissues, but their role in ischemic stroke is unknown. We investigated neoepitope expression in the postischemic brain and the role of natural Abs in recognizing these epitopes and mediating complement-dependent injury. A novel IgM mAb recognizing a subset of phospholipids (C2) and a previously characterized anti-annexin IV mAb (B4) were used to reconstitute and characterize injury in Ab-deficient Rag1(-/-) mice after 60 min of middle cerebral artery occlusion and reperfusion. Reconstitution with C2 or B4 mAb in otherwise protected Rag1(-/-) mice restored injury to that seen in wild-type (wt) mice, as demonstrated by infarct volume, demyelination, and neurologic scoring. IgM deposition was demonstrated in both wt mice and reconstituted Rag1(-/-) mice, and IgM colocalized with the complement activation fragment C3d following B4 mAb reconstitution. Further, recombinant annexin IV significantly reduced infarct volumes in wt mice and in Rag1(-/-) mice administered normal mouse serum, demonstrating that a single Ab reactivity is sufficient to develop cerebral ischemia reperfusion injury in the context of an entire natural Ab repertoire. Finally, C2 and B4 mAbs bound to hypoxic, but not normoxic, human endothelial cells in vitro. Thus, the binding of pathogenic natural IgM to postischemic neoepitopes initiates complement-dependent injury following murine cerebral ischemia and reperfusion, and, based also on previous data investigating IgM reactivity in human serum, there appears to be a similar recognition system in both mouse and man.


Assuntos
Anticorpos Monoclonais/toxicidade , Autoanticorpos , Isquemia Encefálica/imunologia , Traumatismo por Reperfusão/imunologia , Acidente Vascular Cerebral/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Isquemia Encefálica/patologia , Ativação do Complemento , Complemento C3 , Imunoglobulina M , Camundongos , Traumatismo por Reperfusão/etiologia , Acidente Vascular Cerebral/patologia
14.
J Immunol ; 189(9): 4640-7, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23028050

RESUMO

There is mounting evidence indicating an important role for complement in the pathogenesis of cerebral ischemia-reperfusion injury, or ischemic stroke. The role of the alternative complement pathway in ischemic stroke has not been investigated, and there is conflicting data on the role of the terminal pathway. In this study, we show that compared with wild-type mice, mice deficient in the alternative pathway protein factor B or mice treated with the alternative pathway inhibitor CR2-fH have improved outcomes after 60-min middle cerebral artery occlusion and 24-h reperfusion. Factor B-deficient or CR2-fH-treated mice were protected in terms of improved neurologic function and reduced cerebral infarct, demyelination, P-selectin expression, neutrophil infiltration, and microthrombi formation. Mice deficient in both the classical and lectin pathways (C1q/MBL deficient) were also protected from cerebral ischemia-reperfusion injury, and there was no detectable C3d deposition in the ipsilateral brain of these mice. These data demonstrate that the alternative pathway is not alone sufficient to initiate complement activation and indicate that the alternative pathway propagates cerebral injury via amplification of the cascade. Deficiency of C6, a component of the terminal cytolytic membrane attack complex, had no effect on outcome after ischemic stroke, indicating that the membrane attack complex is not involved in mediating injury in this model. We additionally show that the protective effect of factor B deficiency and CR2-fH treatment is sustained in the subacute stage of infarct development, adding to the clinical relevance of these findings.


Assuntos
Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Via Alternativa do Complemento/imunologia , Mediadores da Inflamação/fisiologia , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia , Animais , Isquemia Encefálica/genética , Antígenos CD59/genética , Complemento C1q/deficiência , Complemento C1q/genética , Complemento C6/deficiência , Complemento C6/genética , Fator B do Complemento/deficiência , Fator B do Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/deficiência , Complexo de Ataque à Membrana do Sistema Complemento/genética , Via Alternativa do Complemento/genética , Lectina de Ligação a Manose da Via do Complemento/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão/genética , Acidente Vascular Cerebral/genética
15.
Cereb Circ Cogn Behav ; 4: 100161, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36741272

RESUMO

Introduction: Various lifestyle factors such as chronic hypertension and a high-sodium diet regimen are shown to impact cerebrovascular morphology and structure. Unusual cerebrovascular morphological and structural changes may contribute to cerebral hypoperfusion in Alzheimer's disease (AD). The objective of this study was to examine whether a high-sodium diet mediates cerebrovascular morphology and cerebral perfusion alterations in AD. Methods: Double transgenic mice harboring Aß precursor protein (APPswe) and presenilin-1 (PSEN1) along with wild-type controls were divided into four groups. Group A (APP/PS1) and B (controls) were both fed a high-sodium (4.00%), while group C (APP/PS1) and D (controls) were both fed a low-sodium (0.08% a regular chow diet) for three months. Then, changes in regional cerebral perfusion and diffusion, cerebrovascular morphology, and structure were quantified. Results: A 3-month high-sodium diet causes pyknosis and deep staining in hippocampal neurons and reduced vascular density in both hippocampal and cortical areas (p <0.001) of APP/PS1. Despite vascular density changes, cerebral perfusion was not increased markedly (p = 0.3) in this group, though it was increased more in wild-type controls (p = 0.022). Conclusion: A high-sodium diet regimen causes cerebrovascular morphology alteration in APP/PS1 mouse model of AD.

16.
Res Sq ; 2023 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-37720021

RESUMO

Serum amyloid A (SAA) proteins increase dramatically in the blood following inflammation. Recently, SAAs are increased in humans following stroke and in ischemic animal models. However, the impact of SAAs on whether this signal is critical in the ischemic brain remains unknown. Therefore, we investigated the role of SAA and SAA signaling in the ischemic brain. Wildtype and SAA deficient mice were exposed to middle cerebral artery occlusion and reperfusion, examined for the impact of infarct volumes, behavioral changes, inflammatory markers, TUNEL staining, and BBB changes. The underlying mechanisms were investigated using SAA deficient mice, transgenic mice and viral vectors. SAA levels were significantly increase following MCAo and mice deficient in SAAs showed reduced infarct volumes and improved behavioral outcomes. SAA deficient mice showed a reduction in TUNEL staining, inflammation and decreased glial activation. Mice lacking acute phase SAAs demonstrated a reduction in expression of the NLRP3 inflammasome and SAA/NLRP3 KO mice showed improvement. Restoration of SAA expression via SAA tg mice or adenoviral expression reestablished the detrimental effects of SAA. A reduction in BBB permeability was seen in the SAA KO mice and anti-SAA antibody treatment reduced the effects on ischemic injury. SAA signaling plays a critical role in regulating NLRP3-induced inflammation and glial activation in the ischemic brain. Blocking this signal will be a promising approach for treating ischemic stroke.

17.
J Biol Chem ; 286(6): 4644-58, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21148554

RESUMO

Ceramides, which are membrane sphingolipids and key mediators of cell-stress responses, are generated by a family of (dihydro) ceramide synthases (Lass1-6/CerS1-6). Here, we report that brain development features significant increases in sphingomyelin, sphingosine, and most ceramide species. In contrast, C(16:0)-ceramide was gradually reduced and CerS6 was down-regulated in mitochondria, thereby implicating CerS6 as a primary ceramide synthase generating C(16:0)-ceramide. Investigations into the role of CerS6 in mitochondria revealed that ceramide synthase down-regulation is associated with dramatically decreased mitochondrial Ca(2+)-loading capacity, which could be rescued by addition of ceramide. Selective CerS6 complexing with the inner membrane component of the mitochondrial permeability transition pore was detected by immunoprecipitation. This suggests that CerS6-generated ceramide could prevent mitochondrial permeability transition pore opening, leading to increased Ca(2+) accumulation in the mitochondrial matrix. We examined the effect of high CerS6 expression on cell survival in primary oligodendrocyte (OL) precursor cells, which undergo apoptotic cell death during early postnatal brain development. Exposure of OLs to glutamate resulted in apoptosis that was prevented by inhibitors of de novo ceramide biosynthesis, myriocin and fumonisin B1. Knockdown of CerS6 with siRNA reduced glutamate-triggered OL apoptosis, whereas knockdown of CerS5 had no effect: the pro-apoptotic role of CerS6 was not stimulus-specific. Knockdown of CerS6 with siRNA improved cell survival in response to nerve growth factor-induced OL apoptosis. Also, blocking mitochondrial Ca(2+) uptake or decreasing Ca(2+)-dependent protease calpain activity with specific inhibitors prevented OL apoptosis. Finally, knocking down CerS6 decreased calpain activation. Thus, our data suggest a novel role for CerS6 in the regulation of both mitochondrial Ca(2+) homeostasis and calpain, which appears to be important in OL apoptosis during brain development.


Assuntos
Apoptose/fisiologia , Encéfalo/enzimologia , Cálcio/metabolismo , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/enzimologia , Esfingosina N-Aciltransferase/metabolismo , Células-Tronco/enzimologia , Animais , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Calpaína/genética , Calpaína/metabolismo , Células Cultivadas , Feminino , Técnicas de Silenciamento de Genes , Homeostase/fisiologia , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas do Tecido Nervoso/genética , Oligodendroglia/citologia , Ratos , Ratos Sprague-Dawley , Esfingosina N-Aciltransferase/genética , Células-Tronco/citologia
18.
Proc Natl Acad Sci U S A ; 106(4): 1285-90, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19164583

RESUMO

Glucagon-like peptide-1 (GLP-1) is an endogenous insulinotropic peptide secreted from the gastrointestinal tract in response to food intake. It enhances pancreatic islet beta-cell proliferation and glucose-dependent insulin secretion, and lowers blood glucose and food intake in patients with type 2 diabetes mellitus (T2DM). A long-acting GLP-1 receptor (GLP-1R) agonist, exendin-4 (Ex-4), is the first of this new class of antihyperglycemia drugs approved to treat T2DM. GLP-1Rs are coupled to the cAMP second messenger pathway and, along with pancreatic cells, are expressed within the nervous system of rodents and humans, where receptor activation elicits neurotrophic actions. We detected GLP-1R mRNA expression in both cultured embryonic primary cerebral cortical and ventral mesencephalic (dopaminergic) neurons. These cells are vulnerable to hypoxia- and 6-hydroxydopamine-induced cell death, respectively. We found that GLP-1 and Ex-4 conferred protection in these cells, but not in cells from Glp1r knockout (-/-) mice. Administration of Ex-4 reduced brain damage and improved functional outcome in a transient middle cerebral artery occlusion stroke model. Ex-4 treatment also protected dopaminergic neurons against degeneration, preserved dopamine levels, and improved motor function in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD). Our findings demonstrate that Ex-4 can protect neurons against metabolic and oxidative insults, and they provide preclinical support for the therapeutic potential for Ex-4 in the treatment of stroke and PD.


Assuntos
Citoproteção , Dopamina/metabolismo , Neurônios/patologia , Doença de Parkinson/patologia , Receptores de Glucagon/metabolismo , Acidente Vascular Cerebral/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Infarto Encefálico/tratamento farmacológico , Infarto Encefálico/patologia , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Embrião de Mamíferos/citologia , Exenatida , Regulação da Expressão Gênica/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Mesencéfalo/citologia , Camundongos , Neurônios/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Ratos , Receptores de Glucagon/genética , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Resultado do Tratamento , Peçonhas/farmacologia , Peçonhas/uso terapêutico
19.
FEBS Lett ; 596(22): 2914-2927, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35971617

RESUMO

Previous studies have shown that amyloid-ß oligomers (AßO) bind with high affinity to cellular prion protein (PrPC ). The AßO-PrPC complex binds to cell-surface co-receptors, including the laminin receptor (67LR). Our current studies revealed that in Neuroscreen-1 cells, 67LR is the major co-receptor involved in the cellular uptake of AßO and AßΟ-induced cell death. Both pharmacological (dibutyryl-cAMP, forskolin and rolipram) and physiological (pituitary adenylate cyclase-activating polypeptide) cAMP-elevating agents decreased cell-surface PrPC and 67LR, thereby attenuating the uptake of AßO and the resultant neuronal cell death. These cAMP protective effects are dependent on protein kinase A, but not dependent on the exchange protein directly activated by cAMP. Conceivably, cAMP protects neuronal cells from AßO-induced cytotoxicity by decreasing cell-surface-associated PrPC and 67LR.


Assuntos
Peptídeos beta-Amiloides , Proteínas PrPC , Peptídeos beta-Amiloides/metabolismo , Proteínas Priônicas , Proteínas PrPC/metabolismo , Laminina/metabolismo , Morte Celular , Receptores de Laminina/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase
20.
J Biol Chem ; 285(27): 20675-82, 2010 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20424167

RESUMO

Ca(2+)/calmodulin (CaM)-dependent protein kinase II (CaMKII) is a major mediator of physiological glutamate signaling involved in higher brain functions. Here, we show CaMKII involvement in pathological glutamate signaling relevant in stroke. The novel inhibitor tatCN21 was neuroprotective even when added hours after glutamate insults. By contrast, the "traditional" inhibitor KN93 attenuated excitotoxicity only when present during the insult. Both inhibitors efficiently blocked Ca(2+)/CaM-stimulated CaMKII activity, CaMKII interaction with NR2B and aggregation of CaMKII holoenzymes. However, only tatCN21 but not KN93 blocked the Ca(2+)-independent "autonomous" activity generated by Thr-286 autophosphorylation, the hallmark feature of CaMKII regulation. Mutational analysis further validated autonomous CaMKII activity as the drug target crucial for post-insult neuroprotection. Overexpression of CaMKII wild type but not the autonomy-deficient T286A mutant significantly increased glutamate-induced neuronal death. Maybe most importantly, tatCN21 also significantly reduced infarct size in a mouse stroke model (middle cerebral arterial occlusion) when injected (1 mg/kg intravenously) 1 h after onset of arterial occlusion. Together, these data demonstrate that inhibition of autonomous CaMKII activity provides a promising therapeutic avenue for post-insult neuro-protection after stroke.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fármacos Neuroprotetores/farmacologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Benzilaminas/farmacologia , Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Calmodulina/farmacologia , Morte Celular , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Regulação Enzimológica da Expressão Gênica , Produtos do Gene tat/química , Produtos do Gene tat/farmacologia , Hipocampo/citologia , Hipocampo/fisiologia , Camundongos , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fragmentos de Peptídeos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/fisiologia , Acidente Vascular Cerebral/tratamento farmacológico , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA