Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
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
3.
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.

4.
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
5.
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
6.
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
7.
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
8.
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.

9.
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
10.
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
11.
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
12.
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.

13.
Biochim Biophys Acta ; 1843(3): 640-51, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24333653

RESUMO

Poly(ADP-ribose) polymerase-1 (PARP-1) and its cleavage products regulate cell viability and NF-kappaB activity when expressed in neurons. PARP-1 cleavage generates a 24 kDa (PARP-1(24)) and an 89 kDa fragment (PARP-1(89)). Compared to WT (PARP-1WT), the expression of an uncleavable PARP-1 (PARP-1(UNCL)) or of PARP-1(24) conferred protection from oxygen/glucose deprivation (OGD) or OGD/restoration of oxygen and glucose (ROG) damage in vitro, whereas expression of PARP-1(89) was cytotoxic. Viability experiments were performed in SH-SY5Y, a human neuroblastoma cell line, as well as in rat primary cortical neurons. Following OGD, the higher viability in the presence of PARP-1UNCL or PARP-1(24) was not accompanied with decreased formation of poly(ADP-riboses) or higher NAD levels. PARP-1 is a known cofactor for NF-kappaB, hence we investigated whether PARP-1 cleavage influences the inflammatory response. All PARP-1 constructs mimicked PARP-1WT in regard to induction of NF-kappaB translocation into the nucleus and its increased activation during ischemic challenge. However, expression of PARP-1(89) construct induced significantly higher NF-kB activity than PARP-1WT; and the same was true for NF-kappaB-dependent iNOS promoter binding activity. At a protein level, PARP-1UNCL and PARP-1(24) decreased iNOS (and lower levels of iNOS transcript) and COX-2, and increased Bcl-xL The increased levels of NF-kB and iNOS transcriptional activities, seen with cytotoxic PARP-189, were accompanied by higher protein expression of COX-2 and iNOS (and higher levels of INOS transcript) and lower protein expression of Bcl-xL Taken together, these findings suggest that PARP-1 cleavage products may regulate cellular viability and inflammatory responses in opposing ways during in vitro models of "ischemia".


Assuntos
NF-kappa B/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Caspases/metabolismo , Morte Celular/fisiologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Ciclo-Oxigenase 2/metabolismo , Humanos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
14.
Neuromolecular Med ; 15(4): 771-81, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23775726

RESUMO

Hibernating squirrels slow blood flow to a crawl, but sustain no damage to brain or other tissues. This phenomenon provides an excellent model of natural tolerance to ischemia. Small ubiquitin-like modifier (SUMO) is a 100-residue peptide that modifies other proteins by being attached to the epsilon amino group of specific lysine residues. The discovery of massive SUMOylation (by both SUMO-1 and SUMO-2/3) occurring in the brains of 13-lined ground squirrels (Ictidomys tridecemlineatus) during hibernation torpor had opened the door to the studies on SUMO and ischemic tolerance reviewed here. Ischemic stress was shown to increase the levels of SUMO conjugation, especially SUMO-2/3, mostly during reperfusion in animal models and during restoration of oxygen and glucose in cell culture systems. Over-expression or depletion of SUMOs and/or Ubc9 (the SUMO E2 conjugating enzyme) increases or decreases (respectively) the levels of SUMO conjugates. Elevated global SUMO conjugations were shown to cytoprotect from ischemic insults; conversely, depressed SUMOylation sensitized cells. Global protein conjugation not only by SUMOs, but also by other ubiquitin-like modifiers (ULMs) including NEDD8, ISG15, UFM1 and FUB1 was shown to be significantly increased in the brains of hibernating ground squirrels during torpor. These increases in multiple ULM conjugations may orchestrate the cellular events in hibernating ground squirrels that induce a state of natural tolerance through their multipronged effects. Certain miRNAs such as the miR-200 family and the miR-182 family were shown, at least partly, to control the levels of these ULM conjugations. Lowering the levels of these miRNAs leads to an increase in global SUMOylation/ULM conjugation, thereby providing the tolerance to ischemia. This suggests that these miRNAs may be good targets for therapeutic intervention in stroke.


Assuntos
Isquemia Encefálica/fisiopatologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Sumoilação/fisiologia , Animais , Células Cultivadas , Cisteína Endopeptidases/fisiologia , Modelos Animais de Doenças , Glucose/metabolismo , Hibernação/fisiologia , Humanos , Hipotermia/fisiopatologia , Hipotermia Induzida , Isquemia/fisiopatologia , Camundongos , Camundongos Transgênicos , MicroRNAs/fisiologia , Modelos Animais , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Estresse Oxidativo , Ratos , Traumatismo por Reperfusão/fisiopatologia , Traumatismo por Reperfusão/prevenção & controle , Sciuridae/fisiologia , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/fisiologia , Ubiquitinas/fisiologia
15.
J Neurosci Methods ; 213(1): 1-5, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23174093

RESUMO

The benefits as well as mechanisms of hypothermia in brain injuries are actively studied at the bench and in the clinic. However, methods used in controlling hypothermia vary among laboratories, and usually brain temperatures are not monitored directly in animals due to the need for an invasive procedure. Here we show a method, water immersion technique, which we developed recently to regulate body temperature in mice during hypothermia process. This method significantly reduced the temperature variation around target temperature. Importantly, this method demonstrated a parallel and consistent relationship between rectal temperature and brain temperature (the brain temperature was consistently 0.5C higher than rectal temperature) throughout hypothermia maintenance. This technique may be well adapted to hypothermia studies in mice and other rodents, especially to the assessment and regulation of brain temperature during studies.


Assuntos
Temperatura Corporal/fisiologia , Encéfalo/fisiologia , Hipotermia Induzida/métodos , Anestesia , Animais , Feminino , Gelo , Imersão , Masculino , Camundongos , Reto/fisiologia
16.
PLoS One ; 7(10): e47787, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23094087

RESUMO

Hibernation torpor provides an excellent model of natural tolerance to ischemia. We have previously shown that massive global SUMOylation occurs during hibernation torpor in ground squirrels. We have also shown that overexpression of Ubc9, SUMO-1, or SUMO-2/3 provides protection against ischemic damage in cell lines and cortical neurons exposed to oxygen/glucose deprivation, and in mice exposed to middle cerebral artery occlusion. We have now extended our study to other Ubiquitin-Like-Modifiers (ULMs), which have multiple cellular functions during stress, in order to assess the possibility that they also have roles in tolerance to ischemia. We found that not only SUMO conjugation, but also global protein conjugation by other ULMs including NEDD8, ISG15, UFM1 and FUB1 were significantly increased in the brains of hibernating ground squirrels during torpor. By means of miRNA microarrays of ground squirrel brain samples (from active and torpor phase) we found that the miR-200 family (miR-200a,b,c/miR-141/miR-429) and the miR-182 family (miR-182/miR-183/miR-96) were among the most consistently depressed miRNAs in the brain during the torpor phase as compared to active animals. In addition, we showed that these miRNAs are involved in the expression of various ULM proteins and their global conjugation to proteins. We observed that inhibition of the miR-200 family and/or miR-182 family miRNA activities in SHSY5Y cells increases global protein conjugation by the above ULMs and makes these cells more tolerant to OGD-induced cell death. This is the first report to describe that the natural tolerance to brain ischemia in hibernators is linked to regulation by microRNAs of a broad range of ubiquitin-like modifiers.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica , Hibernação/fisiologia , MicroRNAs/genética , Sciuridae/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Linhagem Celular , Feminino , Glucose/deficiência , Masculino , MicroRNAs/metabolismo , Modelos Biológicos , Oxigênio/metabolismo , Sciuridae/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
17.
PLoS One ; 6(10): e25852, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22016779

RESUMO

We have previously shown that a massive increase in global SUMOylation occurs during torpor in ground squirrels, and that overexpression of Ubc9 and/or SUMO-1 in cell lines and cortical neurons protects against oxygen and glucose deprivation. To examine whether increased global SUMOylation protects against ischemic brain damage, we have generated transgenic mice in which Ubc9 is expressed strongly in all tissues under the chicken ß-actin promoter. Ubc9 expression levels in 10 founder lines ranged from 2 to 30 times the endogenous level, and lines that expressed Ubc9 at modestly increased levels showed robust resistance to brain ischemia compared to wild type mice. The infarction size was inversely correlated with the Ubc9 expression levels for up to five times the endogenous level. Although further increases showed no additional benefit, the Ubc9 expression level was highly correlated with global SUMO-1 conjugation levels (and SUMO-2,3 levels to a lesser extent) up to a five-fold Ubc9 increase. Most importantly, there were striking reciprocal relationships between SUMO-1 (and SUMO-2,3) conjugation levels and cerebral infarction volumes among all tested animals, suggesting that the limit in cytoprotection by global SUMOylation remains undefined. These results support efforts to further augment global protein SUMOylation in brain ischemia.


Assuntos
Encéfalo/metabolismo , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/metabolismo , Sumoilação , Enzimas de Conjugação de Ubiquitina/genética , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Feminino , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , Modelos Lineares , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Sumoilação/genética , Fatores de Tempo , Enzimas de Conjugação de Ubiquitina/metabolismo
18.
J Neurochem ; 109(1): 257-67, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19200349

RESUMO

Ground squirrels in hibernation torpor have been shown to have striking increases in global SUMOylation on tissue immunoblots. Here, we find evidence that global SUMOylation is also involved in ischemic tolerance in primary cortical neuronal cultures (from rats and mice) and SHSY5Y human neuroblastoma cells. Cultured cortical neurons preconditioned by sublethal oxygen/glucose deprivation (OGD) were less vulnerable to severe OGD than non-preconditioned neurons. Preconditioned neurons maintained elevated SUMO-1 conjugation levels (and, to a lesser extent those of SUMO-2/3) on western blots in contrast to non-preconditioned cells. Further, cortical neurons and SHSY5Y cells in which transfected SUMO-1 or SUMO-2 were over-expressed showed increased survival after severe OGD. In contrast, cell cultures subjected to depletion of endogenous SUMO-1 protein by RNAi had reduced survival after exposure to this form of in vitro ischemia and an attenuated protective response to preconditioning. These findings suggest that maintenance of a globally elevated SUMO-1 (and maybe SUMO-2/3) conjugation level as revealed by immunoblot assays is a component of ischemic tolerance.


Assuntos
Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Proteína SUMO-1/metabolismo , Animais , Isquemia Encefálica/patologia , Morte Celular/fisiologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Precondicionamento Isquêmico , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley
19.
J Cereb Blood Flow Metab ; 27(5): 950-62, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-16955077

RESUMO

Hibernation torpor provides an excellent natural model of tolerance to profound reductions in blood flow to the brain and other organs. Here, we report that during torpor of 13-lined ground squirrels, massive SUMOylation occurs in the brain, liver, and kidney. The level of small ubiquitin-related modifier (SUMO) conjugation coincides with the expression level of Ubc9, the SUMO specific E2-conjugating enzyme. Hypothermia alone also increased SUMO conjugation, but not as markedly as hibernation torpor. Increased SUMO conjugation (induced by Ubc9 overexpression, ischemic preconditioning (PC)+/-hypothermia) was necessary and sufficient for tolerance of SHSY5Y neuroblastoma cells to oxygen/glucose deprivation (OGD) ('in vitro ischemia'); decreased SUMO conjugation (induced by a dominant-negative Ubc9) severely reduced tolerance to OGD in these cells. These data indicate that post-translational modification of proteins by SUMOylation is a prominent feature of hibernation torpor and is critical for cytoprotection by ischemic PC+/-hypothermia in SHSY5Y cells subjected to OGD.


Assuntos
Hibernação/fisiologia , Hipotermia/patologia , Precondicionamento Isquêmico , Proteínas do Tecido Nervoso/metabolismo , Sciuridae/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Benzimidazóis , Western Blotting , Peso Corporal/fisiologia , Morte Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/fisiologia , Células Cultivadas , Clonagem Molecular , Feminino , Citometria de Fluxo , Corantes Fluorescentes , Glucose/deficiência , Hipóxia Encefálica/fisiopatologia , Imuno-Histoquímica , Masculino , Microscopia Confocal , Propídio , Transfecção
20.
J Biol Chem ; 280(42): 35742-50, 2005 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-16115883

RESUMO

Mitochondria are important participants in apoptosis, releasing cytochrome c into the cytoplasm and undergoing extensive fragmentation. However, mechanisms underlying these processes remain unclear. Here, we demonstrate that cytochrome c release during apoptosis precedes mitochondrial fragmentation. Unexpectedly, OPA1, a dynamin-like GTPase of the mitochondrial intermembrane space important for maintaining cristae structure, is co-released with cytochrome c. To mimic the loss of OPA1 occurring after its release, we knocked down OPA1 expression using RNA interference. This triggered structural changes in the mitochondrial cristae and caused increased fragmentation by blocking mitochondrial fusion. Because cytochrome c is mostly sequestered within cristae folds but released rapidly and completely during apoptosis, we examined the effect of OPA1 loss on cytochrome c release, demonstrating that it is accelerated. Thus, our results suggest that an initial mitochondrial leak of OPA1 leads to cristae structural alterations and exposure of previously sequestered protein pools, permitting continued release in a feed-forward manner to completion. Moreover, our findings indicate that the resulting OPA1 depletion causes a block in mitochondrial fusion, providing a compelling mechanism for the prominent increase in mitochondrial fragmentation seen during apoptosis.


Assuntos
Apoptose , Citocromos c/metabolismo , GTP Fosfo-Hidrolases/fisiologia , Mitocôndrias/metabolismo , Carbono/metabolismo , Citosol/metabolismo , Relação Dose-Resposta a Droga , Endopeptidase K/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Cinética , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Cloreto de Potássio/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA