Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
bioRxiv ; 2023 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-37205496

RESUMO

Ischemic stroke results in a loss of tissue homeostasis and integrity, the underlying pathobiology of which stems primarily from the depletion of cellular energy stores and perturbation of available metabolites 1 . Hibernation in thirteen-lined ground squirrels (TLGS), Ictidomys tridecemlineatus , provides a natural model of ischemic tolerance as these mammals undergo prolonged periods of critically low cerebral blood flow without evidence of central nervous system (CNS) damage 2 . Studying the complex interplay of genes and metabolites that unfolds during hibernation may provide novel insights into key regulators of cellular homeostasis during brain ischemia. Herein, we interrogated the molecular profiles of TLGS brains at different time points within the hibernation cycle via RNA sequencing coupled with untargeted metabolomics. We demonstrate that hibernation in TLGS leads to major changes in the expression of genes involved in oxidative phosphorylation and this is correlated with an accumulation of the tricarboxylic acid (TCA) cycle intermediates citrate, cis-aconitate, and α-ketoglutarate-αKG. Integration of the gene expression and metabolomics datasets led to the identification of succinate dehydrogenase (SDH) as the critical enzyme during hibernation, uncovering a break in the TCA cycle at that level. Accordingly, the SDH inhibitor dimethyl malonate (DMM) was able to rescue the effects of hypoxia on human neuronal cells in vitro and in mice subjected to permanent ischemic stroke in vivo . Our findings indicate that studying the regulation of the controlled metabolic depression that occurs in hibernating mammals may lead to novel therapeutic approaches capable of increasing ischemic tolerance in the CNS.

2.
Curr Issues Mol Biol ; 35: 127-144, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31422937

RESUMO

Brain ischaemia is a severe form of metabolic stress that activates a cascade of pathological events involving many signalling pathways. Modulation of these pathways is largely mediated by post-translational modifications (PTMs). Indeed, PTMs can rapidly modify pre-existing proteins by attaching chemical or polypeptide moieties to selected amino acid residues, altering their functions, stability, subcellular localizations, or interactions with other proteins. Subsequently, related signalling pathways can be substantially affected. Thus, PTMs are widely deployed by cells as an adaptive strategy at the front line to efficiently cope with internal and external stresses. Many types of PTMs have been identified, including phosphorylation, O-GlcNAcylation, small ubiquitin-like modifier (SUMO) modification (SUMOylation), and ubiquitination. All these PTMs have been studied in brain ischaemia to some extent. In particular, a large body of evidence has demonstrated that both global SUMOylation and ubiquitination are massively activated after brain ischaemia, and this activation may play a critical role in defining the fate and function of cells in the post-ischaemic brain. The goal of this review will be to summarize the current findings on SUMOylation and ubiquitination in brain ischaemia and discuss their clinical implications.


Assuntos
Isquemia Encefálica/enzimologia , Isquemia Encefálica/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Ubiquitinação , Animais , Isquemia Encefálica/patologia , Isquemia Encefálica/terapia , Linhagem Celular , Humanos , Proteoma/genética , Proteoma/metabolismo , Transdução de Sinais/genética
3.
Ann Clin Transl Neurol ; 6(4): 817-820, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31020007

RESUMO

We aimed to characterize peripheral blood gene expression profile of penumbra defined as MRI perfusion-diffusion mismatch (PD MM) in peripheral blood of patients with acute ischemic stroke. We studied 23 patients. Perfusion-diffusion mismatch volume was observed to be associated and significantly correlated with the expression of 34 genes including those related to inflammation, SUMOylation, and coagulation; while lipopolysaccharide inhibition was identified to be a candidate upstream regulator of these processes (z-score -2.38, P = 0.04). Penumbral volume is correlated with a specific gene expression profile in the peripheral blood characterized by overlap of inflammatory and neuroprotective pathways that are regulated by lipopolysaccharide inhibition.


Assuntos
Isquemia Encefálica/genética , Encéfalo/patologia , Circulação Cerebrovascular/fisiologia , Acidente Vascular Cerebral/genética , Idoso , Idoso de 80 Anos ou mais , Encéfalo/metabolismo , Isquemia Encefálica/diagnóstico , Imagem de Difusão por Ressonância Magnética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos de Pesquisa , Acidente Vascular Cerebral/diagnóstico
4.
EBioMedicine ; 42: 214-224, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30905846

RESUMO

BACKGROUND: Neural stem cell (NSC)-based therapies hold great promise for treating diseases of the central nervous system (CNS). However, several fundamental problems still need to be overcome to fully exploit the clinical potential of NSC therapeutics. Chief among them is the limited survival of NSC grafts within hostile microenvironments. METHODS: Herein, we sought to engineer NSCs in an effort to increase graft survival within ischemic brain lesions via upregulation of global SUMOylation, a post-translational modification critically involved in mediating tolerance to ischemia/reperfusion. FINDINGS: NSCs overexpressing the SUMO E2-conjugase Ubc9 displayed resistance to oxygen-glucose-deprivation/restoration of oxygen/glucose (OGD/ROG) and enhanced neuronal differentiation in vitro, as well as increased survival and neuronal differentiation when transplanted in mice with transient middle cerebral artery occlusion in vivo. INTERPRETATION: Our work highlights a critical role for SUMOylation in NSC biology and identifies a biological pathway that can be targeted to increase the effectiveness of exogenous stem cell medicines in ischemic stroke. FUND: Intramural Research Program of the NINDS/NIH, the Italian Multiple Sclerosis Foundation (FISM), the Bascule Charitable Trust, NIH-IRTA-OxCam and Wellcome Trust Research Training Fellowships.


Assuntos
Sobrevivência Celular , Células-Tronco Neurais/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Biomarcadores , Ciclo Celular/genética , Sobrevivência Celular/genética , Biologia Computacional/métodos , Metabolismo Energético , Expressão Gênica , Perfilação da Expressão Gênica , Glucose/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Oxigênio/metabolismo , Transdução de Sinais , Transplante de Células-Tronco , Acidente Vascular Cerebral/etiologia , Sumoilação , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
5.
Chromosome Res ; 26(4): 307-315, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30443803

RESUMO

Сonstitutive heterochromatin areas are revealed by differential staining as C-positive chromosomal regions. These C-positive bands may greatly vary by location, size, and nucleotide composition. CBG-banding is the most commonly used method to detect structural heterochromatin in animals. The difficulty in identification of individual chromosomes represents an unresolved problem of this method as the body of the chromosome is stained uniformly and does not have banding pattern beyond C-bands. Here, we present the method that we called CDAG for sequential heterochromatin staining after differential GTG-banding. The method uses G-banding followed by heat denaturation in the presence of formamide with consecutive fluorochrome staining. The new technique is valid for the concurrent revealing of heterochromatin position due to differential banding of chromosomes and heterochromatin composition (AT-/GC-rich) in animal karyotyping.


Assuntos
Bandeamento Cromossômico/métodos , Heterocromatina/química , Animais , Composição de Bases , Corantes Fluorescentes , Formamidas/farmacologia , Cariotipagem , Desnaturação de Ácido Nucleico , Coloração e Rotulagem
7.
Lab Invest ; 98(6): 799-813, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29472640

RESUMO

The intestinal epithelium constitutes a crucial defense to the potentially life-threatening effects of gut microbiota. However, due to a complex underlying vasculature, hypoperfusion and resultant tissue ischemia pose a particular risk to function and integrity of the epithelium. The small ubiquitin-like modifier (SUMO) conjugation pathway critically regulates adaptive responses to metabolic stress and is of particular significance in the gut, as inducible knockout of the SUMO-conjugating enzyme Ubc9 results in rapid intestinal epithelial disintegration. Here we analyzed the pattern of individual SUMO isoforms in intestinal epithelium and investigated their roles in intestinal ischemia/reperfusion (I/R) damage. Immunostaining revealed that epithelial SUMO2/3 expression was almost exclusively limited to crypt epithelial nuclei in unchallenged mice. However, intestinal I/R or overexpression of Ubc9 caused a remarkable enhancement of epithelial SUMO2/3 staining along the crypt-villus axis. Unexpectedly, a similar pattern was found in SUMO1 knockout mice. Ubc9 transgenic mice, but also SUMO1 knockout mice were protected from I/R injury as evidenced by better preserved barrier function and blunted inflammatory responses. PCR array analysis of microdissected villus-tip epithelia revealed a specific epithelial contribution to reduced inflammatory responses in Ubc9 transgenic mice, as key chemotactic signaling molecules such as IL17A were significantly downregulated. Together, our data indicate a critical role particularly of the SUMO2/3 isoforms in modulating responses to I/R and provide the first evidence that SUMO1 deletion activates a compensatory process that protects from ischemic damage.


Assuntos
Mucosa Intestinal/irrigação sanguínea , Traumatismo por Reperfusão/prevenção & controle , Proteína SUMO-1/fisiologia , Enzimas de Conjugação de Ubiquitina/fisiologia , Animais , Quimiocinas/análise , Mucosa Intestinal/química , Microdissecção e Captura a Laser , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína SUMO-1/deficiência , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/análise , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Enzimas de Conjugação de Ubiquitina/genética , Ubiquitinas/análise , Ubiquitinas/fisiologia
8.
Cell Stem Cell ; 22(3): 355-368.e13, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29478844

RESUMO

Neural stem cell (NSC) transplantation can influence immune responses and suppress inflammation in the CNS. Metabolites, such as succinate, modulate the phenotype and function of immune cells, but whether and how NSCs are also activated by such immunometabolites to control immunoreactivity and inflammatory responses is unclear. Here, we show that transplanted somatic and directly induced NSCs ameliorate chronic CNS inflammation by reducing succinate levels in the cerebrospinal fluid, thereby decreasing mononuclear phagocyte (MP) infiltration and secondary CNS damage. Inflammatory MPs release succinate, which activates succinate receptor 1 (SUCNR1)/GPR91 on NSCs, leading them to secrete prostaglandin E2 and scavenge extracellular succinate with consequential anti-inflammatory effects. Thus, our work reveals an unexpected role for the succinate-SUCNR1 axis in somatic and directly induced NSCs, which controls the response of stem cells to inflammatory metabolic signals released by type 1 MPs in the chronically inflamed brain.


Assuntos
Sistema Nervoso Central/patologia , Inflamação/patologia , Macrófagos/metabolismo , Células-Tronco Neurais/citologia , Ácido Succínico/metabolismo , Animais , Linhagem Celular , Doença Crônica , Dinoprostona/metabolismo , Feminino , Humanos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/transplante , Fosforilação Oxidativa , Receptores Acoplados a Proteínas G/metabolismo , Ácido Succínico/líquido cefalorraquidiano
9.
Front Mol Neurosci ; 11: 22, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29440989

RESUMO

Hibernating 13-lined ground squirrels (Ictidomys tridecemlineatus; TLGS) rank among the most brain hypoperfusion-tolerant mammals known. Herein we provide some evidence of cycling between an epithelial phenotype and a hybrid epithelial/mesenchymal (E/M) phenotype (partial EMT) within the brains of TLGS during each bout of hibernation torpor. During hibernation torpor, expression of the epithelial marker E-cadherin (E-CDH) was reduced, while expression of the well-known mesenchymal markers vimentin and Sox2 were increased. P-cadherin (P-CDH), which has recently been proposed as a marker of intermediate/partial EMT, also increased during torpor, suggesting that a partial EMT may be taking place during hibernation torpor. Members of the miR-200 family and miR-182 cluster and Akt isoforms (Akt1, Akt2), well-known EMT regulators, were also differentially regulated in the TLGS brain during hibernation bouts. Using SHSY5Y cells, we also demonstrate that the Akt1/Akt2 ratio determined the expression levels of miR-200/miR-182 miRNA family members, and that these miRNAs controlled the expression of EMT-related proteins. Accordingly, we propose that such cell state transitions (EMT/MET) may be one of the mechanisms underlying the extraordinary ischemic tolerance of the TLGS brain during hibernation bouts; hibernator brain cells appear to enter reversible states that confer the stress survival characteristics of cancer cells without the risk of neoplastic transformation.

10.
J Neurosci Res ; 96(4): 487-500, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28463430

RESUMO

Traumatic brain injuries (TBIs) pose a massive burden of disease and continue to be a leading cause of morbidity and mortality throughout the world. A major obstacle in developing effective treatments is the lack of comprehensive understanding of the underlying mechanisms that mediate tissue damage and recovery after TBI. As such, our work aims to highlight the development of a novel experimental platform capable of fully characterizing the underlying pathobiology that unfolds after TBI. This platform encompasses an empirically optimized multiplex immunohistochemistry staining and imaging system customized to screen for a myriad of biomarkers required to comprehensively evaluate the extent of neuroinflammation, neural tissue damage, and repair in response to TBI. Herein, we demonstrate that our multiplex biomarker screening platform is capable of evaluating changes in both the topographical location and functional states of resident and infiltrating cell types that play a role in neuropathology after controlled cortical impact injury to the brain in male Sprague-Dawley rats. Our results demonstrate that our multiplex biomarker screening platform lays the groundwork for the comprehensive characterization of changes that occur within the brain after TBI. Such work may ultimately lead to the understanding of the governing pathobiology of TBI, thereby fostering the development of novel therapeutic interventions tailored to produce optimal tissue protection, repair, and/or regeneration with minimal side effects, and may ultimately find utility in a wide variety of other neurological injuries, diseases, and disorders that share components of TBI pathobiology.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Imuno-Histoquímica/métodos , Neuroimagem/métodos , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/metabolismo , Encéfalo/fisiopatologia , Lesões Encefálicas Traumáticas/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Masculino , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Ratos Sprague-Dawley , Doenças da Língua/metabolismo , Doenças da Língua/patologia
11.
FASEB J ; 32(3): 1677-1691, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29146736

RESUMO

The development of novel neuroprotective treatments for acute stroke has been fraught with failures, which supports the view of ischemic brain damage as a highly complex multifactorial process. Post-translational modifications such as small ubiquitin-like modifier (SUMO)ylation have emerged as critical molecular regulatory mechanisms in states of both homeostasis and ischemic stress, as evidenced by our previous work. Accordingly, the clinical significance of the selective control of the global SUMOylation process has become apparent in studies of ischemic pathobiology and pathophysiology. Herein, we describe a process capable of identifying and characterizing small molecules with the potential of targeting the SUMO system through inhibition of SUMO deconjugation in an effort to develop novel stroke therapies.-Bernstock, J. D., Ye, D., Smith, J. A., Lee, Y.-J., Gessler, F. A., Yasgar, A., Kouznetsova, J., Jadhav, A., Wang, Z., Pluchino, S., Zheng, W., Simeonov, A., Hallenbeck, J. M., Yang, W. Quantitative high-throughput screening identifies cytoprotective molecules that enhance SUMO-conjugation via the inhibition of SUMO-specific protease (SENP)2.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cisteína Endopeptidases/metabolismo , Inibidores de Proteases/farmacologia , Proteína SUMO-1/metabolismo , Sumoilação , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Transformada , Cisteína Endopeptidases/genética , Humanos , Ratos , Proteína SUMO-1/genética , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
12.
J Cereb Blood Flow Metab ; 38(1): 5-16, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29148315

RESUMO

Post-translational protein modification by small ubiquitin-like modifier (SUMO) regulates a myriad of homeostatic and stress responses. The SUMOylation pathway has been extensively studied in brain ischemia. Convincing evidence is now at hand to support the notion that a major increase in levels of SUMOylated proteins is capable of inducing tolerance to ischemic stress. Therefore, the SUMOylation pathway has emerged as a promising therapeutic target for neuroprotection in the face of brain ischemia. Despite this, it is prudent to acknowledge that there are many key questions still to be addressed in brain ischemia related to SUMOylation. Accordingly, herein, we provide a critical review of literature within the field to summarize current knowledge and in so doing highlight pertinent translational implications of the SUMOylation pathway in brain ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Neuroproteção/fisiologia , Sumoilação/fisiologia , Humanos
13.
Sci Rep ; 7(1): 7425, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28785061

RESUMO

Protein SUMOylation is a dynamic post-translational modification shown to be involved in a diverse set of physiologic processes throughout the cell. SUMOylation has also been shown to play a role in the pathobiology of myriad cancers, one of which is glioblastoma multiforme (GBM). As such, the clinical significance and therapeutic utility offered via the selective control of global SUMOylation is readily apparent. There are, however, relatively few known/effective inhibitors of global SUMO-conjugation. Herein we describe the identification of topotecan as a novel inhibitor of global SUMOylation. We also provide evidence that inhibition of SUMOylation by topotecan is associated with reduced levels of CDK6 and HIF-1α, as well as pronounced changes in cell cycle progression and cellular metabolism, thereby highlighting its putative role as an adjuvant therapy in defined GBM patient populations.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Glioblastoma/patologia , Sumoilação/efeitos dos fármacos , Topotecan/farmacologia , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quinase 6 Dependente de Ciclina/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Células Tumorais Cultivadas
14.
J Cereb Blood Flow Metab ; 37(7): 2314-2319, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28303738

RESUMO

Ischemic stroke continues to be a leading cause of morbidity and mortality throughout the world. To protect and/or repair the ischemic brain, a multitiered approach may be centered on neural stem cell (NSC) transplantation. Transplanted NSCs exert beneficial effects not only via structural replacement, but also via immunomodulatory and/or neurotrophic actions. Unfortunately, the clinical translation of such promising therapies remains elusive, in part due to their limited persistence/survivability within the hostile ischemic microenvironment. Herein, we discuss current approaches for the development of NSCs more amenable to survival within the ischemic brain as a tool for future cellular therapies in stroke.


Assuntos
Isquemia Encefálica/terapia , Engenharia Celular/métodos , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Acidente Vascular Cerebral/terapia , Condicionamento Pré-Transplante/métodos , Isquemia Encefálica/complicações , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Sobrevivência Celular , Humanos , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/patologia , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia
15.
Front Neurol ; 8: 738, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29375471

RESUMO

Posttranslational modification by small ubiquitin-like modifier (SUMO) regulates myriad physiological processes within cells and has been demonstrated to be highly activated in murine brains after cerebral ischemia. Numerous in vitro and murine in vivo studies have demonstrated that this increased SUMO conjugation is an endogenous neuroprotective stress response that has potential in being leveraged to develop novel therapies for ischemic stroke. However, SUMO activation has not yet been studied in poststroke human brains, presenting a clear limitation in translating experimental successes in murine models to human patients. Accordingly, here, we present a case wherein the brain tissue of a stroke patient (procured shortly after death) was processed by multiplex immunohistochemistry to investigate SUMO activation.

16.
J Neurochem ; 138(1): 101-16, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27087120

RESUMO

The putative neuroprotective properties of various flavonoids have long been reported. Among this class of chemicals, quercetin, a major flavone/flavonol naturally occurring in plants, deserves focused attention because of the myriad of beneficial effects observed in various in vitro and in vivo models of central nervous system damage/degeneration. However, the mechanisms governing the beneficial outcomes mediated by quercetin remain to be elucidated. In an effort to define the underlying molecular mechanisms, our study employed human/rat neuroblastoma cell lines (SHSY5Y and B35, respectively) and E18-derived rat primary cortical neurons upon which the effects of various flavonoids were examined. Of note, increases in the levels of global SUMOylation, a post-translational modification with the Small Ubiquitin-like MOdifier (SUMO) were pronounced. Quercetin treatment increased SUMOylation levels in both SHSY5Y cells and rat cortical neurons in a dose and time-dependent manner, possibly via the direct inactivation of certain SENPs (SUMO-specific isopeptidases). Of particular interest, cells treated with quercetin displayed increased tolerance to oxygen/glucose deprivation exposure, an in vitro model of ischemia. SHSY5Y cells treated with quercetin also increased the expression of Nrf2 (via a decrease in the levels of Keap1), heme oxygenase-1 (HO-1), and nitric oxide synthase 1 (NOS1), which provide further protection from oxidative stress. In addition, the increased SUMOylation of HIF-1α was noted and deemed to be significant. We hypothesize that SUMOylated HIF-1α plays a fundamental role in the protection afforded and may underlie some of quercetin's ability to protect cells from oxygen/glucose deprivation-induced cell death, via an up-regulation of HO-1 and NOS1, which ultimately leads to the induction of pro-life NOS1/protein kinase G signaling. Quercetin acts to increase survival in the face of ischemia via an increase of SENP3 expression, the possible inactivation of SENPs 1/2, and via a decrease in KEAP1 levels (thereby increasing Nrf2 stability). These changes may then lead to increase in HIF-1α SUMOylation and HO-1 activation, followed by an up-regulation of NOS1/PKG signaling. Pathways altered via quercetin treatment within our experimental system are represented by blue arrowheads. Solid black arrows represent relationships that have been explored while a dotted arrow represents a relationship that has yet to be confirmed.


Assuntos
Glucose/deficiência , Hipóxia/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Quercetina/farmacologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Córtex Cerebral/citologia , Feminino , Humanos , L-Lactato Desidrogenase/metabolismo , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Gravidez , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
17.
J Cereb Blood Flow Metab ; 36(2): 426-41, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26661196

RESUMO

The conjugation/de-conjugation of Small Ubiquitin-like Modifier (SUMO) has been shown to be associated with a diverse set of physiologic/pathologic conditions. The clinical significance and ostensible therapeutic utility offered via the selective control of the global SUMOylation process has become readily apparent in ischemic pathophysiology. Herein, we describe the development of a novel quantitative high-throughput screening (qHTS) system designed to identify small molecules capable of increasing SUMOylation via the regulation/inhibition of members of the microRNA (miRNA)-182 family. This assay employs a SHSY5Y human neuroblastoma cell line stably transfected with a dual firefly-Renilla luciferase reporter system for identification of specific inhibitors of either miR-182 or miR-183. In this study, we have identified small molecules capable of inducing increased global conjugation of SUMO in both SHSY5Y cells and rat E18-derived primary cortical neurons. The protective effects of a number of the identified compounds were confirmed via an in vitro ischemic model (oxygen/glucose deprivation). Of note, this assay can be easily repurposed to allow high-throughput analyses of the potential drugability of other relevant miRNA(s) in ischemic pathobiology.


Assuntos
Glucose/deficiência , Hipóxia/patologia , MicroRNAs/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Sumoilação/efeitos dos fármacos , Animais , Córtex Cerebral/efeitos dos fármacos , Conjugação Genética/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Cultura Primária de Células , Ratos , Bibliotecas de Moléculas Pequenas , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/efeitos dos fármacos , Pesquisa Translacional Biomédica/métodos
18.
PLoS One ; 10(10): e0140772, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26473731

RESUMO

The immune system plays a fundamental role in both the development and pathobiology of stroke. Inflammasomes are multiprotein complexes that have come to be recognized as critical players in the inflammation that ultimately contributes to stroke severity. Inflammasomes recognize microbial and host-derived danger signals and activate caspase-1, which in turn controls the production of the pro-inflammatory cytokine IL-1ß. We have shown that A151, a synthetic oligodeoxynucleotide containing multiple telemeric TTAGGG motifs, reduces IL-1ß production by activated bone marrow derived macrophages that have been subjected to oxygen-glucose deprivation and LPS stimulation. Further, we demonstrate that A151 reduces the maturation of caspase-1 and IL-1ß, the levels of both the iNOS and NLRP3 proteins, and the depolarization of mitochondrial membrane potential within such cells. In addition, we have demonstrated that A151 reduces ischemic brain damage and NLRP3 mRNA levels in SHR-SP rats that have undergone permanent middle cerebral artery occlusion. These findings clearly suggest that the modulation of inflammasome activity via A151 may contribute to a reduction in pro-inflammatory cytokine production by macrophages subjected to conditions that model brain ischemia and modulate ischemic brain damage in an animal model of stroke. Therefore, modulation of ischemic pathobiology by A151 may have a role in the development of novel stroke prevention and therapeutic strategies.


Assuntos
Isquemia Encefálica/imunologia , Glucose/imunologia , Inflamassomos/imunologia , Macrófagos/imunologia , Motivos de Nucleotídeos , Oligodesoxirribonucleotídeos/farmacologia , Oxigênio/imunologia , Telômero , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Caspase 1/imunologia , Caspase 1/metabolismo , Glucose/metabolismo , Inflamassomos/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Oxigênio/metabolismo , Ratos , Ratos Endogâmicos SHR
19.
J Neurochem ; 135(5): 943-57, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26375300

RESUMO

Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) have an extraordinary capacity to withstand prolonged and profound reductions in blood flow and oxygen delivery to the brain without incurring any cellular damage. As such, the hibernation torpor of I. tridecemlineatus provides a valuable model of tolerance to ischemic stress. Herein, we report that during hibernation torpor, a marked reduction in the phosphorylation of the ribosomal protein S6 (rpS6) occurs within the brains of I. tridecemlineatus. Of note, rpS6 phosphorylation was shown to increase in the brains of rats that underwent an occlusion of the middle cerebral artery. However, such an increase was attenuated after the implementation of an ischemic preconditioning paradigm. In addition, cultured cortical neurons treated with the rpS6 kinase (S6K) inhibitors, D-glucosamine or PF4708671, displayed a decrease in rpS6 phosphorylation and a subsequent increase in tolerance to oxygen/glucose deprivation, an in vitro model of ischemic stroke. Collectively, such evidence suggests that the down-regulation of rpS6 signal transduction may account for a substantial part of the observed increase in cellular tolerance to brain ischemia that occurs during hibernation torpor and after ischemic preconditioning. Further identification and characterization of the mechanisms used by hibernating species to increase ischemic tolerance may eventually clarify how the loss of homeostatic control that occurs during and after cerebral ischemia in the clinic can ultimately be minimized and/or prevented. Mammalian hibernation provides a valuable model of tolerance to ischemic stress. Herein, we demonstrate that marked reductions in the phosphorylation of ribosomal protein S6 (rpS6), extracellular signal-regulated kinase family of mitogen-activated protein (MAP) kinase p44/42 (p44/42MAPK) and ribosomal protein S6 kinase (S6K) occur within the brains of both hibernating squirrels and rats, which have undergone an ischemic preconditioning paradigm. We therefore propose that the down-regulation of rpS6 signal transduction may account for a substantial part of the observed increase in cellular tolerance to brain ischemia that occurs during hibernation torpor and after ischemic preconditioning, via a suppression of protein synthesis and/or energy consumption.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hibernação , Infarto da Artéria Cerebral Média/prevenção & controle , Precondicionamento Isquêmico , Proteína S6 Ribossômica/metabolismo , Animais , Células Cultivadas , Córtex Cerebral , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/genética , Glucose/deficiência , Masculino , Dados de Sequência Molecular , Neurônios/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteína S6 Ribossômica/genética , Sciuridae , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Tempo
20.
Front Cell Neurosci ; 8: 416, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25538566

RESUMO

The molecular mechanisms underlying hypothermic neuroprotection have yet to be fully elucidated. Herein we demonstrate that global SUMOylation, a form of post-translational modification with the Small Ubiquitin-like MOdifer, participates in the multimodal molecular induction of hypothermia-induced ischemic tolerance. Mild (32°C) to moderate (28°C) hypothermic treatment(s) during OGD (oxygen-glucose-deprivation) or ROG (restoration of oxygen/glucose) increased global SUMO-conjugation levels and protected cells (both SHSY5Y and E18 rat cortical neurons) from OGD and ROG-induced cell death. Hypothermic exposure either before or after permanent middle cerebral artery occlusion (pMCAO) surgery in wild type mice increased global SUMO-conjugation levels in the brain and in so doing protected these animals from pMCAO-induced ischemic damage. Of note, hypothermic exposure did not provide an additional increase in protection from pMCAO-induced ischemic brain damage in Ubc9 transgenic (Ubc9 Tg) mice, which overexpress the sole E2 SUMO conjugating enzyme and thereby display elevated basal levels of global SUMOylation under normothermic conditions. Such evidence suggests that increases in global SUMOylation are critical and may account for a substantial part of the observed increase in cellular tolerance to brain ischemia caused via hypothermia.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...