Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Int J Mol Sci ; 24(12)2023 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-37373475

RESUMO

Recent studies show that repetitive transcranial magnetic stimulation (rTMS) improves cognitive and motor functions in patients with Parkinson's Disease (PD). Gamma rhythm low-field magnetic stimulation (LFMS) is a new non-invasive rTMS technique that generates diffused and low-intensity magnetic stimulation to the deep cortical and subcortical areas. To investigate the potential therapeutic effects of LFMS in PD, we subjected an experimental mouse model to LFMS (as an early treatment). We examined the LFMS effect on motor functions as well as neuronal and glial activities in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated male C57BL/6J mice. Mice received MPTP injection (30 mg/kg, i.p., once daily for 5 days) followed by LFMS treatment, 20 min each day for 7 days. LFMS treatment improved motor functions compared with the sham-treated MPTP mice. Further, LFMS significantly improved tyrosine hydroxylase (TH) and decreased glial fibrillary acidic protein (GFAP) levels in substantia nigra pars compacta (SNpc) and non-significantly in striatal (ST) regions. LFMS treatment improved neuronal nuclei (NeuN) levels in SNpc. Our findings suggest that early LFMS treatment improves neuronal survival and, in turn, motor functions in MPTP-treated mice. Further investigation is required to clearly define the molecular mechanisms by which LFMS improves motor and cognitive function in PD patients.


Assuntos
Dopamina , Doença de Parkinson , Masculino , Animais , Camundongos , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Substância Negra/metabolismo , Camundongos Endogâmicos C57BL , Doença de Parkinson/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Fenômenos Magnéticos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Modelos Animais de Doenças
2.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445708

RESUMO

Brain injury/concussion is a growing epidemic throughout the world. Although evidence supports association between traumatic brain injury (TBI) and disturbance in brain glucose metabolism, the underlying molecular mechanisms are not well established. Previously, we reported the release of cellular prion protein (PrPc) from the brain to circulation following TBI. The PrPc level was also found to be decreased in insulin-resistant rat brains. In the present study, we investigated the molecular link between PrPc and brain insulin resistance in a single and repeated mild TBI-induced mouse model. Mild TBI was induced in mice by dropping a weight (~95 g at 1 m high) on the right side of the head. The procedure was performed once and thrice (once daily) for single (SI) and repeated induction (RI), respectively. Micro PET/CT imaging revealed that RI mice showed significant reduction in cortical, hippocampal and cerebellum glucose uptake compared to SI and control. Mice that received RI also showed significant motor and cognitive deficits. In co-immunoprecipitation, the interaction between PrPc, flotillin and Cbl-associated protein (CAP) observed in the control mice brains was disrupted by RI. Lipid raft isolation showed decreased levels of PrPc, flotillin and CAP in the RI mice brains. Based on observation, it is clear that PrPc has an interaction with CAP and the dislodgment of PrPc from cell membranes may lead to brain insulin resistance in a mild TBI mouse model. The present study generated a new insight into the pathogenesis of brain injury, which may result in the development of novel therapy.


Assuntos
Concussão Encefálica/fisiopatologia , Resistência à Insulina/fisiologia , Animais , Encéfalo/metabolismo , Concussão Encefálica/diagnóstico por imagem , Lesões Encefálicas/complicações , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Glucose/metabolismo , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tomografia por Emissão de Pósitrons/métodos , Proteínas Priônicas/metabolismo , Príons/metabolismo , Transdução de Sinais/fisiologia
3.
Mol Brain ; 14(1): 89, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099009

RESUMO

The type 1 and type 2 cannabinoid receptors (CB1 and CB2 receptors) are class A G protein-coupled receptors (GPCRs) that are activated by endogenous lipids called endocannabinoids to modulate neuronal excitability and synaptic transmission in neurons throughout the central nervous system (CNS), and inflammatory processes throughout the body. CB1 receptor is one of the most abundant GPCRs in the CNS and is involved in many physiological and pathophysiological processes, including mood, appetite, and nociception. CB2 receptor is primarily found on immunomodulatory cells of both the CNS and the peripheral immune system. In this study, we isolated lipid raft and non-lipid raft fractions of plasma membrane (PM) from mouse cortical tissue by using cold non-ionic detergent and sucrose gradient centrifugation to study the localization of CB1 receptor and CB2 receptor. Lipid raft and non-lipid raft fractions were confirmed by flotillin-1, caveolin-1 and transferrin receptor as their protein biomarkers. Both CB1 receptor and CB2 receptor were found in non-raft compartments that is inconsistent with previous findings in cultured cell lines. This study demonstrates compartmentalization of both CB1 receptor and CB2 receptor in cortical tissue and warrants further investigation of CB1 receptor and CB2 receptor compartmental distribution in various brain regions and cell types.


Assuntos
Membrana Celular/metabolismo , Córtex Cerebral/metabolismo , Receptores de Canabinoides/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Microdomínios da Membrana/metabolismo , Camundongos Endogâmicos C57BL
4.
Brain Inj ; 35(6): 734-741, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33760683

RESUMO

BACKGROUND: Cellular prion protein (PrPC) is a lipid raft protein abundant within CNS. It is regulated by a disintegrin and metalloproteinase domain containing protein 10 (ADAM10). PrPC has previously been implicated as a biomarker for TBI. ADAM10 has not been investigated as a TBI biomarker. OBJECTIVE: We evaluated PrPC and ADAM10 as candidate biomarkers for TBI. METHODS: We performed ELISA for ADAM10 and PrPC on plasma samples of patients with TBI admitted to Brigham and Women's Hospital. Plasma samples from 20 patients admitted for isolated TBI were acquired from a biobank with clinical information. Control plasma (37 samples) was acquired from a commercial source. GraphPad was used to conduct statistical analysis. RESULTS: 37 controls and 20 TBI samples were collected. Of the patients with TBI, eight were mild, three were moderate, and nine were severe. Both PrPC and ADAM10 were elevated in patients with TBI compared with control (p < .001). ADAM10 exhibited greater expression in patients with worse clinical grade. There was no significant association of either PrPC or ADAM10 with time after injury. CONCLUSIONS: Our results indicate that PrPC and ADAM10 appear to be useful potential tools for screening of TBI. ADAM10 is closely associated with clinical grade.


Assuntos
Lesões Encefálicas Traumáticas , Príons , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide , Biomarcadores , Feminino , Humanos , Proteínas de Membrana , Projetos Piloto , Proteínas Priônicas
5.
Int J Mol Sci ; 21(19)2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32992764

RESUMO

Cellular prion protein (PrPc) is a small glycosylphosphatidylinositol (GPI) anchored protein most abundantly found in the outer leaflet of the plasma membrane (PM) in the central nervous system (CNS). PrPc misfolding causes neurodegenerative prion diseases in the CNS. PrPc interacts with a wide range of protein partners because of the intrinsically disordered nature of the protein's N-terminus. Numerous studies have attempted to decipher the physiological role of the prion protein by searching for proteins which interact with PrPc. Biochemical characteristics and biological functions both appear to be affected by interacting protein partners. The key challenge in identifying a potential interacting partner is to demonstrate that binding to a specific ligand is necessary for cellular physiological function or malfunction. In this review, we have summarized the intracellular and extracellular interacting partners of PrPc and potential consequences of their binding. We also briefly describe prion disease-related mutations at the end of this review.


Assuntos
Doenças Priônicas/metabolismo , Proteínas Priônicas/metabolismo , Animais , Humanos , Ligantes , Proteínas Priônicas/genética
6.
Front Neurol ; 11: 833, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973652

RESUMO

Epilepsy comprises more than 40 clinical syndromes affecting millions of patients and families worldwide. To decode the molecular and pathological framework of epilepsy researchers, need reliable human epilepsy and control brain samples. Brain bank organizations collecting and supplying well-documented clinically and pathophysiologically tissue specimens are important for high-quality neurophysiology and neuropharmacology studies for epilepsy and other neurological diseases. New development in molecular mechanism and new treatment methods for neurological disorders have evoked increased demands for human brain tissue. An epilepsy brain bank is a storage source for both the frozen samples as well as the formaldehyde fixed paraffin embedded (FFPE) tissue from epilepsy surgery resections. In 2014, the University of Saskatchewan have started collecting human epilepsy brain tissues for the first time in Canada. This review highlights the necessity and importance of Epilepsy Brain bank that provides unique access for research to valuable source of brain tissue and blood samples from epilepsy patients.

7.
Clin Exp Pharmacol Physiol ; 47(9): 1530-1536, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32304254

RESUMO

Insulin-mediated signalling in the brain is critical for neuronal functioning. Insulin resistance is implicated in the development of some neurological diseases, although changes associated with absence epilepsy have not been established yet. Therefore, we examined the major components of PI3K/Akt-mediated insulin signalling in cortical, thalamic, and hippocampal tissues collected from Genetic Absence Epilepsy Rats from Strasbourg (GAERS) and Non-Epileptic Control (NEC) rats. Insulin levels were also measured in plasma and cerebrospinal fluid (CSF). For the brain samples, the nuclear fraction (NF) and total homogenate (TH) were isolated and investigated for insulin signalling markers including insulin receptor beta (IRß), IR substrate-1 and 2 (IRS1 & 2), phosphatase and tensin homologue (PTEN), phosphoinositide 3-kinase phospho-85 alpha (PI3K p85α), phosphatidylinositol 4,5-bisphosphate, phosphatidylinositol (3,4,5)-trisphosphate, protein kinase B (PKB/Akt1/2/3), glucose transporter-1 and 4 (GLUT1 & 4) and glycogen synthase kinase-3ß (GSK3ß) using western blotting. A significant increase in PTEN and GSK3ß levels and decreased PI3K p85α and pAkt1/2/3 levels were observed in NF of GAERS cortical and hippocampal tissues. IRß, IRS1, GLUT1, and GLUT4 levels were significantly decreased in hippocampal TH of GAERS compared to NEC. A non-significant increase in insulin levels was observed in plasma and CSF of GAERS rats. An insulin sensitivity assay showed decreased p-Akt level in cortical and hippocampal tissues. Together, altered hippocampal insulin signalling was more prominent in NF and TH compared to cortical and thalamic regions in GAERS. Restoring insulin signalling may improve the pathophysiology displayed by GAERS, including the spike-and-wave discharges that relate to absence seizures in patients.


Assuntos
Ondas Encefálicas , Epilepsia Tipo Ausência/metabolismo , Insulina/metabolismo , Rombencéfalo/metabolismo , Animais , Glicemia/metabolismo , Modelos Animais de Doenças , Epilepsia Tipo Ausência/genética , Epilepsia Tipo Ausência/fisiopatologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Insulina/sangue , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Endogâmicos , Receptor de Insulina/metabolismo , Rombencéfalo/fisiopatologia , Transdução de Sinais
8.
Neurosci Lett ; 716: 134641, 2020 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-31759082

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has been well documented in glycolytic pathway. Independent of this, it has various other functions including stimulator of programmed cell death. Reports suggest that glutamate receptor AMPA type-2 subunit (GluA2) forms protein complex with GAPDH and internalized during excitotoxicity. Further, nuclear accumulation of GluA2 and GAPDH have been studied in neurological disorders like epilepsy and multiple sclerosis, and disruption of this complex rescued neurological symptoms such as astrogliosis, AMPA mediated excitotoxicity and p53 phosphorylation. On the other hand, study on ischemic rat model showed that nucleus translocated GAPDH binds with p53 leading to apoptosis. However, the molecular events underlying these processes remained to be established in Parkinson's disease (PD). The present study focused on investigating the levels of GAPDH, GluA2 and p53 in the nuclear fraction (NF) and total homogenate (TH) of substantia nigral (SN) region obtained from post-mortem PD brains and their age-matched controls. The level of caspase 3, an apoptotic marker and mediator for p53 induced cell death was also measured. A significant increase in nuclear GAPDH, GluA2 and p53 were observed in PD SN region, compared to the controls. Similarly, increased caspase 3 level was observed in PD SN region. Data obtained from the present study suggest that nuclear accumulation of GAPDH, GluA2 and p53 plays a key role in the pathophysiology of neuronal cell death in PD. Thus decreasing nuclear translocation of these death pro-death signaling markers may attenuate neurodegeneration that aids in the development of potential therapeutic targets in the management of PD.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Doença de Parkinson/fisiopatologia , Receptores de AMPA/metabolismo , Substância Negra/fisiopatologia , Proteína Supressora de Tumor p53/metabolismo , Idoso , Autopsia , Núcleo Celular/metabolismo , Feminino , Humanos , Masculino , Doença de Parkinson/metabolismo , Substância Negra/metabolismo
9.
J Neurotrauma ; 36(22): 3103-3114, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31020907

RESUMO

Traumatic brain injury (TBI)/concussion is a growing epidemic throughout the world. Memory and neurobehavioral dysfunctions are among the sequelae of TBI. Dislodgement of cellular prion protein (PrPc) and disruption of circadian rhythm have been linked to TBI. Low-field magnetic stimulation (LFMS) is a new noninvasive repetitive transcranial magnetic stimulation (rTMS) technique that generates diffused and low-intensity magnetic stimulation to deep cortical and subcortical areas. The role of LFMS on PrPc, proteins related to the circadian rhythm, and behavior alterations in a repeated TBI mouse model were studied in the present study. TBI was induced to the mice (right hemisphere) using weight-drop method, once daily for 3 days. LFMS treatment was given for 20 min once daily for 4 days (immediately after each TBI induction). The results showed that LFMS-treated TBI mice significantly improved cognitive and motor function as evidenced by open field exploration, rotarod, and novel location recognition tasks. In addition, a significant increase in PrPc and decreased glial fibrillary acidic protein levels were observed in cortical and hippocampal regions of LFMS-treated TBI mice brain compared with sham-treated TBI mice, while neuronal nuclei level was significantly increased in cortical region. In LFMS-treated mice, a decrease in proteins related to circadian rhythm were observed, compared with sham-treated TBI mice. The results obtained from the study demonstrated the neuroprotective effect of LFMS, which may be through regulating PrPc and/or proteins related to circadian rhythm. Thus, the present study suggests that LFMS may improve the subject's neurological condition following TBI.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Proteínas Priônicas/efeitos da radiação , Recuperação de Função Fisiológica/efeitos da radiação , Estimulação Magnética Transcraniana/métodos , Animais , Lesões Encefálicas Traumáticas/metabolismo , Ritmo Circadiano/efeitos da radiação , Cognição/efeitos da radiação , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos da radiação , Proteínas Priônicas/metabolismo
10.
Mol Neurobiol ; 56(4): 2508-2523, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30039334

RESUMO

Pilocarpine-induced status epilepticus (SE), which results in the development of spontaneous recurrent seizures (SRSs) activates glutamatergic receptors that contribute to seizure sustenance and neuronal cell death. In the current study, we evaluate whether the exposure to perampanel, an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor blocker, or amantadine, a N-methyl-D-aspartic acid (NMDA) receptor blocker would reduce the SE-induced long-term consequences. SE was induced in adult male Sprague Dawley rats with pilocarpine. Perampanel or amantadine was injected 10 or 60 min after SE onset. The efficacy of either, in overcoming pilocarpine-induced SE was assessed using electroencephalogram (EEG) recordings. In addition, alterations in cognitive function, development of spontaneous recurrent seizures (SRSs), and hippocampal damage that are generally encountered after SE were also assessed at 72 h and 5 weeks after the induction of SE. Our results indicate that both early and late treatment with perampanel but not amantadine significantly reduced seizure activity. Furthermore, perampanel but not amantadine, reversed the memory deficits in Y-maze and novel object recognition (NOR) tests and retarded the appearance of SRSs. Moreover, perampanel treatment led to reduced SE-induced caspase-3 activation in the hippocampal lysates. Taken together, the data obtained from the study reveals that blocking AMPA receptors by perampanel can modify SE-induced long-term consequences. Our results may provide a proof of principle for the potential therapeutic application of perampanel in clinical use for status epilepticus in future.


Assuntos
Amantadina/uso terapêutico , Comportamento Animal , Piridonas/uso terapêutico , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/prevenção & controle , Amantadina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Nitrilas , Pilocarpina , Subunidades Proteicas/metabolismo , Piridonas/farmacologia , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Convulsões/tratamento farmacológico
11.
Eur J Neurosci ; 49(9): 1157-1170, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30554430

RESUMO

Traumatic brain injury (TBI) is a leading major cause of morbidity and mortality in youth and individuals under 45 year age. A wide variety of cellular and molecular mechanisms have been identified contributing to the pathogenesis of TBI. A better understanding of the pathophysiology behind TBI is essential for providing more effective treatment. Excitotoxicity as one of the secondary molecular events is a major contributing factor in apoptosis and neuronal death following the initial injury in TBI. Excitotoxicity is the rapid overload and influx of calcium into the cell cytoplasm, activating a series of deleterious signaling cascades causing the cell to undergo apoptosis. Conventional understanding is that the rapid influx of calcium is initiated through glutamate release. However, there are overlooked glutamate-independent mechanisms that cause the rapid calcium influx into the neuronal cytoplasm, evoking or contributing to excitotoxicity. Therefore, the focus of this review will be on the role of the glutamate-independent excitotoxic mechanisms of the mechanosensitive response of NMDA receptors, mechanoporation of the cell membrane, ischemia, and the release of calcium from intracellular stores. In conclusion, the shear and stretch forces during a TBI event may result in the mechanosensitive activation of NMDA receptors which contribute to glutamate-independent excitotoxicity.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Apoptose/fisiologia , Lesões Encefálicas Traumáticas/metabolismo , Cálcio/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Estresse Mecânico
12.
Neurosci Lett ; 666: 139-143, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29288045

RESUMO

Studies showed that 50-80% of Parkinson's disease (PD) patients have been reported with abnormal glucose tolerance. Alterations in glucose and energy metabolism serve as the early molecular event in PD. Although evidences support that the insulin resistance plays a major role in motor and non-motor complications of PD, the underlying mechanism in the pathogenesis of PD is unclear. To address this issue, we investigated the alterations in major components of insulin signaling in nuclear fraction (NF) and whole tissue homogenate (TH) of substantia nigral (SN) region obtained from postmortem PD brain and their age-matched controls. Pathway components include insulin receptor ß (IRß), IR substrate-1 (IRS1), phosphoinositide 3-kinase p85 (PI3K p85), phosphatidylinositol 4,5-bisphosphate (PIP2), phosphatidylinositol (3,4,5)-trisphosphate (PIP3), protein kinase B (PKB/Akt1/2/3) and glycogen synthase kinase-3ß (GSK3ß). Phosphatase and tensin homolog (PTEN), a negative regulator of insulin signaling cascade was also studied. A significant decrease in nuclear PI3K p85, Akt1/2/3 and PIP3 levels and significant increase in nuclear PTEN and GSK3ß levels were observed in SN region of PD brain when compared to the age-matched controls. Consistently, significant decrease in IRß, IRS1, PI3K p85, Akt1/2/3 and PIP3 levels and increased GSK3ß level were observed in TH obtained from SN region of PD brain compared to the control brain. Data from the study suggest that alterations in insulin signaling may play a vital role in the pathogenesis/progression of PD and other related complications. Thus, decreasing nuclear accumulation of PTEN and/or restoring insulin signaling cascade may halt the neurodegeneration in PD.


Assuntos
Insulina/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Doença de Parkinson/metabolismo , Substância Negra/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais/fisiologia
13.
Epilepsy Res ; 136: 1-4, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28719803

RESUMO

Studies in animal models and human tissues show that nuclear translocation of sterol regulatory element binding protein 1 (SREBP1) and glutamate A2 subunit (GluA2) of cell-surface AMPA receptor (AMPAR) trigger neuronal excitotoxicity-induced apoptosis in stroke. However, it is not known whether a similar type of underlying pathophysiology occurs in absence epilepsy. To explore this issue, we examined the levels of mature SREBP1, GluA2, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), p53, and activated to total caspase 3 ratio in nuclear fractions (NF) of hippocampal homogenate from 8 to 10 week old male Genetic Absence Epilepsy Rats from Strasbourg (GAERS) and non-epileptic control (NEC) strains. Mature SREBP1 and GluA2 levels were elevated approximately two-fold in NFs of GAERS hippocampal homogenates compared to NEC animals. Significant increases in GAPDH (∼15-fold) and total caspase 3 (∼10-fold) levels were also found in NFs of GAERS hippocampal homogenates in comparison to the non-epileptic strain. Data from the current study suggest that absence epilepsy in GAERS is associated with nuclear translocation of mature SREBP1, GluA2 subunit of AMPARs, and recruitment of pro-cell death signaling proteins such as GAPDH and caspase 3. These changes may contribute to hippocampal neuronal/glial cell death in GAERS. Therefore, inhibiting the nuclear accumulation of mature SREBP1 and GluA2 translocation may reduce the pathophysiology of absence epilepsy.


Assuntos
Epilepsia Tipo Ausência/metabolismo , Hipocampo/metabolismo , Receptores de AMPA/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Western Blotting , Caspase 3/metabolismo , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Genes p53 , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Masculino , Técnicas de Transferência Nuclear , Ratos
14.
Biochem Biophys Res Commun ; 488(1): 136-140, 2017 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-28483521

RESUMO

Sterol regulatory element-binding protein1 (SREBP1) is a key regulatory factor that controls lipid homeostasis. Overactivation of SREBP1 and elevated lipid biogenesis are considered the major characteristics in malignancies of prostate cancer, endometrial cancer, and glioblastoma. However, the impact of SREBP1 activation in the progression of pancreatic cancer has not been explored. The present study examines the effect of suppression of SREBP1 activation by its inhibitors like fatostatin and PF429242 besides analyzing the impact of inhibitory effects on SREBP1 downstream signaling cascade such as fatty acid synthase (FAS), hydroxymethylglutaryl-CoA reductase (HMGCoAR), stearoyl-CoA desaturase-1 (SCD-1), and tumor suppressor protein p53 in MIA PaCa-2 pancreatic cancer cells. Both fatostatin and PF429242 inhibited the growth of MIA PaCa-2 cells in a time and concentration-dependent manner with maximal inhibition attained at 72 h time period with IC50 values of 14.5 µM and 24.5 µM respectively. Detailed Western blot analysis performed using fatostatin and PF429242 at 72 h time point led to significant decrease in the levels of the active form of SREBP1 and its downstream signaling proteins such as FAS, SCD-1 and HMGCoAR and the mutant form of tumor suppressor protein, p53, levels in comparison to the levels observed in vehicle treated control group of MIA PaCa-2 pancreatic cells over the same time period. Our in vitro data suggest that SREBP1 may contribute to pancreatic tumor growth and its inhibitors could be considered as a potential target in the management of pancreatic cancer cell proliferation.


Assuntos
Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Piridinas/farmacologia , Pirrolidinas/farmacologia , Proteína de Ligação a Elemento Regulador de Esterol 1/antagonistas & inibidores , Tiazóis/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Neoplasias Pancreáticas/metabolismo , Piridinas/administração & dosagem , Pirrolidinas/administração & dosagem , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Relação Estrutura-Atividade , Tiazóis/administração & dosagem
15.
J Alzheimers Dis ; 58(4): 1003-1016, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28527218

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder of the elderly. As the prevalence of AD rises in the 21st century, there is an urgent need for the development of effective pharmacotherapies. Currently, drug treatments target the symptoms of the disease and do not modify or halt the disease progress. Thus, natural compounds have been investigated for their ability to treat AD. This review examines the efficacy of curcumin, a polyphenol derived from turmeric herb, to treat AD. We summarize the in vivo and in vitro research describing the mechanisms of action in which curcumin modifies AD pathology: curcumin inhibits the formation and promotes the disaggregation of amyloid-ß plaques, attenuates the hyperphosphorylation of tau and enhances its clearance, binds copper, lowers cholesterol, modifies microglial activity, inhibits acetylcholinesterase, mediates the insulin signaling pathway, and is an antioxidant. In conclusion, curcumin has the potential to be more efficacious than current treatments. However, its usefulness as a therapeutic agent may be hindered by its low bioavailability. If the challenge of low bioavailability is overcome, curcumin-based medications for AD may be in the horizon.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Anti-Inflamatórios/uso terapêutico , Curcumina/uso terapêutico , Doença de Alzheimer/metabolismo , Animais , Progressão da Doença , Humanos
16.
PLoS One ; 10(2): e0117286, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25643046

RESUMO

Sport-related mild traumatic brain injury (mTBI) or concussion is a significant health concern to athletes with potential long-term consequences. The diagnosis of sport concussion and return to sport decision making is one of the greatest challenges facing health care clinicians working in sports. Blood biomarkers have recently demonstrated their potential in assisting the detection of brain injury particularly, in those cases with no obvious physical injury. We have recently discovered plasma soluble cellular prion protein (PrP(C)) as a potential reliable biomarker for blast induced TBI (bTBI) in a rodent animal model. In order to explore the application of this novel TBI biomarker to sport-related concussion, we conducted a pilot study at the University of Saskatchewan (U of S) by recruiting athlete and non-athlete 18 to 30 year-old students. Using a modified quantitative ELISA method, we first established normal values for the plasma soluble PrP(C) in male and female students. The measured plasma soluble PrP(C) in confirmed concussion cases demonstrated a significant elevation of this analyte in post-concussion samples. Data collected from our pilot study indicates that the plasma soluble PrP(C) is a potential biomarker for sport-related concussion, which may be further developed into a clinical diagnostic tool to assist clinicians in the assessment of sport concussion and return-to-play decision making.


Assuntos
Traumatismos em Atletas/sangue , Concussão Encefálica/sangue , Proteínas PrPC/sangue , Proteínas PrPC/química , Esportes , Adolescente , Adulto , Biomarcadores/sangue , Biomarcadores/química , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Projetos Piloto , Solubilidade , Adulto Jovem
17.
J Neurotrauma ; 32(1): 58-65, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25058115

RESUMO

Traumatic brain injury (TBI) is deemed the "signature injury" of recent military conflicts in Afghanistan and Iraq, largely because of increased blast exposure. Injuries to the brain can often be misdiagnosed, leading to further complications in the future. Therefore, the use of protein biomarkers for the screening and diagnosis of TBI is urgently needed. In the present study, we have investigated the plasma levels of soluble cellular prion protein (PrPC) as a novel biomarker for the diagnosis of primary blast-induced TBI (bTBI). We hypothesize that the primary blast wave can disrupt the brain and dislodge extracellular localized PrPC, leading to a rise in concentration within the systemic circulation. Adult male Sprague-Dawley rats were exposed to single pulse shockwave overpressures of varying intensities (15-30 psi or 103.4-206.8 kPa] using an advanced blast simulator. Blood plasma was collected 24 h after insult, and PrPC concentration was determined with a modified commercial enzyme-linked immunosorbent assay (ELISA) specific for PrPC. We provide the first report that mean PrPC concentration in primary blast exposed rats (3.97 ng/mL ± 0.13 SE) is significantly increased compared with controls (2.46 ng/mL ± 0.14 SE; two tailed test p < 0.0001). Furthermore, we report a mild positive rank correlation between PrPC concentration and increasing blast intensity (psi) reflecting a plateaued response at higher pressure magnitudes, which may have implications for all military service members exposed to blast events. In conclusion, it appears that plasma levels of PrPC may be a novel biomarker for the detection of primary bTBI.


Assuntos
Traumatismos por Explosões/sangue , Lesões Encefálicas/diagnóstico , Proteínas PrPC/sangue , Animais , Biomarcadores/sangue , Traumatismos por Explosões/complicações , Lesões Encefálicas/sangue , Lesões Encefálicas/etiologia , Explosões , Masculino , Ratos , Ratos Sprague-Dawley
18.
Biochem Biophys Res Commun ; 448(2): 151-6, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24780399

RESUMO

The risk of stroke is drastically increased in diabetic and pre-diabetic patients. The worldwide spread of obesity and insulin resistance increases the incidence of stroke. The direct effect of insulin resistance, as it pertains to stroke, on the central nervous system is not well understood. Since one of the physiological functions of the cellular prion protein (PrP(C)) is neuroprotection, we studied effects of brain insulin resistance on the expression of PrP(C) in fructose-fed rats as an animal model of prediabetes. Compared with control chow-fed animals, rats fed a high-fructose diet (FF), exhibited compromised tyrosine phosphorylation of insulin receptor ß subunit (IRß) and reduced serine phosphorylation of Akt, PI3K activity, and decreased PIP3 levels in cortices indicating the induction of brain insulin resistance. We also observed that both mRNA and protein expression of the PrP(C) was significantly decreased whereas protein level of NR2B subunit of NMDA glutamate receptors profoundly increased in the brain of fructose-fed rats compared to control chow-fed rats. Considering a neuroprotective role for PrP(C) and the involvement of NR2B subunit in the excitotoxicity-induced neuronal apoptosis, these phenomena may contribute to the severity and poor prognosis of ischemic stroke in diabetes/prediabetes.


Assuntos
Complicações do Diabetes/fisiopatologia , Resistência à Insulina/fisiologia , Proteínas PrPC/metabolismo , Estado Pré-Diabético/metabolismo , Acidente Vascular Cerebral/etiologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Complicações do Diabetes/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Frutose/efeitos adversos , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas PrPC/genética , Estado Pré-Diabético/complicações , Estado Pré-Diabético/fisiopatologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Acidente Vascular Cerebral/fisiopatologia
19.
Chem Biol Drug Des ; 83(5): 507-20, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24325540

RESUMO

For decades, biomedical and pharmaceutical researchers have worked to devise new and more effective therapeutics to treat diseases affecting the central nervous system. The blood-brain barrier effectively protects the brain, but poses a profound challenge to drug delivery across this barrier. Many traditional drugs cannot cross the blood-brain barrier in appreciable concentrations, with less than 1% of most drugs reaching the central nervous system, leading to a lack of available treatments for many central nervous system diseases, such as stroke, neurodegenerative disorders, and brain tumors. Due to the ineffective nature of most treatments for central nervous system disorders, the development of novel drug delivery systems is an area of great interest and active research. Multiple novel strategies show promise for effective central nervous system drug delivery, giving potential for more effective and safer therapies in the future. This review outlines several novel drug delivery techniques, including intranasal drug delivery, nanoparticles, drug modifications, convection-enhanced infusion, and ultrasound-mediated drug delivery. It also assesses possible clinical applications, limitations, and examples of current clinical and preclinical research for each of these drug delivery approaches. Improved central nervous system drug delivery is extremely important and will allow for improved treatment of central nervous system diseases, causing improved therapies for those who are affected by central nervous system diseases.


Assuntos
Sistema Nervoso Central/metabolismo , Portadores de Fármacos/química , Barreira Hematoencefálica/metabolismo , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Humanos , Lipídeos/química , Nanopartículas/química , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo
20.
PLoS One ; 8(10): e76860, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146937

RESUMO

Neural stem cell (NSC) replacement therapy is considered a promising cell replacement therapy for various neurodegenerative diseases. However, the low rate of NSC survival and neurogenesis currently limits its clinical potential. Here, we examined if hippocampal long-term potentiation (LTP), one of the most well characterized forms of synaptic plasticity, promotes neurogenesis by facilitating proliferation/survival and neuronal differentiation of NSCs. We found that the induction of hippocampal LTP significantly facilitates proliferation/survival and neuronal differentiation of both endogenous neural progenitor cells (NPCs) and exogenously transplanted NSCs in the hippocampus in rats. These effects were eliminated by preventing LTP induction by pharmacological blockade of the N-methyl-D-aspartate glutamate receptor (NMDAR) via systemic application of the receptor antagonist, 3-[(R)-2-carboxypiperazin-4-yl]-propyl-1-phosphonic acid (CPP). Moreover, using a NPC-neuron co-culture system, we were able to demonstrate that the LTP-promoted NPC neurogenesis is at least in part mediated by a LTP-increased neuronal release of brain-derived neurotrophic factor (BDNF) and its consequent activation of tropomysosin receptor kinase B (TrkB) receptors on NSCs. Our results indicate that LTP promotes the neurogenesis of both endogenous and exogenously transplanted NSCs in the brain. The study suggests that pre-conditioning of the host brain receiving area with a LTP-inducing deep brain stimulation protocol prior to NSC transplantation may increase the likelihood of success of using NSC transplantation as an effective cell therapy for various neurodegenerative diseases.


Assuntos
Diferenciação Celular , Potenciação de Longa Duração/fisiologia , Células-Tronco Neurais/citologia , Neurônios/citologia , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Giro Denteado/citologia , Células HEK293 , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transplante de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...