Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 99: 107901, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34273637

RESUMO

Periodontitis is initiated by serious and sustained bacterial infection and ultimately results in chronic immune-mediated inflammation, tissue destruction, and bone loss. The pathogenesis of periodontitis remains unclear. Host immunological responses to periodontal bacteria ultimately determine the severity and mechanisms governing periodontitis progression. This study aimed to clarify the effect of the hypoxia-inducible factor-1α (HIF-1α) activator dimethyloxalylglycine (DMOG) on a mouse periodontitis model and its underlying role in macrophage polarization. qRT-PCR analysis showed that DMOG inhibited the M1-like polarization of both RAW264.7 macrophages and murine bone marrow macrophages (BMMs) and downregulated TNF-α, IL-6, CD86, and MCP-1 expression in vitro. Immunofluorescence staining and flow cytometry also confirmed the less percentage of F4/80 + CD86 + cells after DMOG treatment. The phosphorylation of NF-κB pathway was also inhibited by DMOG with higher level of HIF-1α expression. Furthermore, mice treated with DMOG showed decreased alveolar bone resorption in the experimental periodontitis model, with significant increases in alveolar bone volume/tissue volume (BV/TV) and bone mineral density (BMD). DMOG treatment of mice decreased the ratio of M1/M2 (CD86+/CD206+) macrophages in periodontal tissues, resulting in the downregulation of proinflammatory cytokines such as TNF-α and IL-6 and increased levels of anti-inflammatory factors such as IL-4 and IL-10. DMOG treatment promoted the number of HIF-1α-positive cells in periodontal tissues. This study demonstrated the cell-specific roles of DMOG in macrophage polarization in vitro and provided insight into the mechanism underlying the protective effect of DMOG in a model of periodontitis.


Assuntos
Perda do Osso Alveolar/tratamento farmacológico , Aminoácidos Dicarboxílicos/uso terapêutico , Macrófagos/efeitos dos fármacos , Periodontite/tratamento farmacológico , Perda do Osso Alveolar/diagnóstico por imagem , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/patologia , Aminoácidos Dicarboxílicos/farmacologia , Animais , Citocinas/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia , Macrófagos/imunologia , Masculino , Maxila/diagnóstico por imagem , Maxila/patologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , Periodontite/diagnóstico por imagem , Periodontite/imunologia , Periodontite/patologia , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Microtomografia por Raio-X
2.
J Neurotrauma ; 36(24): 3394-3409, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31232175

RESUMO

Spinal cord injury (SCI) is a devastating neurological disorder that usually leads to a loss of motor and sensory function in patients. The expression of hypoxia inducible factor-1α (HIF-1α) is increased, and exerts a protective role after traumatic SCI. However, the endogenous activity of HIF-1α is insufficient for promoting functional recovery. The present study tested the potential effect of the sustained activation of HIF-1α by the prolylhydroxylase (PHD) inhibitor dimethyloxalylglycine (DMOG) on anti-apoptotic process and the regulation of axonal regeneration after SCI. Here, we found that treatment with DMOG significantly increased the expression of HIF-1α and that the stabilization of HIF-1α induced by DMOG not only decreased the expression of apoptotic proteins to promote neural survival, but also enhanced axonal regeneration by regulating microtubule stabilization in vivo and in vitro. In addition, we found that DMOG promoted neural survival and axonal regeneration by activating autophagy, which is induced by the HIF-1α/BNIP3 signaling pathway, and that the inhibition of HIF-1α or autophagy abrogated the protective effect of DMOG, as expected. Taken together, our results demonstrate that treatment with DMOG improves functional recovery after SCI and that DMOG may serve as a potential candidate for treating SCI.


Assuntos
Aminoácidos Dicarboxílicos/uso terapêutico , Axônios/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Regeneração Nervosa/fisiologia , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/metabolismo , Aminoácidos Dicarboxílicos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Axônios/efeitos dos fármacos , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Regeneração Nervosa/efeitos dos fármacos , Células PC12 , Estabilidade Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia
3.
Physiol Behav ; 199: 265-272, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30500334

RESUMO

Mesenchymal stem cell (MSC) transplantation therapy has been proposed as a promising approach for the treatment of neurodegenerative disease. Chemical and pharmacological preconditioning before transplantation could optimize the therapeutic properties of transplanted MSCs. In this study, we hypothesized that preconditioning treatment with a prolyl hydroxylase inhibitor, dimethyloxalylglycine (DMOG), will increase MSC efficacy and paracrine effects in an amyloid-ß (Aß)-injected Alzheimer rat model. MSCs were incubated in different concentrations of DMOG for 24 h. Cell viability, migration, and antioxidant capacity was assessed in DMOG-treated and non-treated MSCs before transplantation into Aß-injected rats. In vitro analysis revealed that DMOG treatment increased cell viability, migration, and expression of CXCR4, CCR2, Nrf2, and HIF-1α in the MSCs. Our in vivo results show that DMOG preconditioning enhances a MSC-mediated rescue of learning and memory function in Aß-injected rats. Furthermore, we found an increased level of BDNF and total antioxidant capacity in the hippocampus of Aß-injected rats following transplantation of preconditioned relative to untreated MSCs. Our results suggest that preconditioning MSCs with DMOG before transplantation may enhance the efficacy of stem cell based therapy in neurodegenerative disease.


Assuntos
Doença de Alzheimer/terapia , Aminoácidos Dicarboxílicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Doença de Alzheimer/induzido quimicamente , Aminoácidos Dicarboxílicos/farmacologia , Peptídeos beta-Amiloides , Animais , Modelos Animais de Doenças
4.
BMC Oral Health ; 15: 60, 2015 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-25981588

RESUMO

BACKGROUND: Rapid wound healing of oral soft tissue may reduce the opportunity of infection and discomfort of patients. Previous studies have demonstrated that enhancement of angiogenesis is an effective way to accelerate wound repair. In this study, to enhance angiogenesis and healing of palatal wounds, dimethyloxalylglycine (DMOG) was applied to a rat palatal wound model. DMOG is known to inhibit oxygen-dependent degradation of hypoxia inducible factor-1 alpha (HIF-1α), which can lead to up-regulation of angiogenesis markers, favoring wound repair. We also evaluated the effects of DMOG on cell migration and HIF-1α expression of rat palatal (RP) cells. Furthermore, mRNA and protein expression of vascular endothelial growth factor (VEGF) were analyzed in DMOG-treated RP cells. METHODS: Primary cultures of rat palatal (RP) cells were obtained from Sprague-Dawley (SD) rats. Effects of DMOG on cell viability and migration of RP cells were evaluated by using a formazan and culture insert, respectively. VEGF mRNA was observed by real-time PCR, and VEGF and HIF-1α proteins were detected by Western blotting. For the animal study, excisional wounds, 3 mm in diameter, were made at the central part of the palate of SD rats. DMOG with hyaluronic acid ointment was topically applied three times during 1 week, and then wound closures were quantitated photographically and histologically. RESULTS: DMOG was cytotoxic to RP cells at concentrations higher than 2 mM and did not affect cell migration at non-cytotoxic concentrations. mRNA and protein expression of VEGF were significantly stimulated by DMOG treatment. The protein level of HIF-1α was also stabilized in RP cells by DMOG. In the animal study, groups treated with 1 mg/ml DMOG showed an increase of rat palatal wound contractures. CONCLUSIONS: DMOG enhanced wound healing of rat palatal mucosa, which was likely due to the angiogenic effect of the agent.


Assuntos
Aminoácidos Dicarboxílicos/uso terapêutico , Mucosa Bucal/lesões , Palato/lesões , Proteínas Angiogênicas/farmacologia , Animais , Técnicas de Cultura de Células , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ácido Hialurônico/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Masculino , Modelos Animais , Mucosa Bucal/efeitos dos fármacos , Palato/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Cicatrização/efeitos dos fármacos
5.
Magn Reson Med ; 70(5): 1481-90, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23280475

RESUMO

MRI is a method of choice for assessing anatomical structures or angiogenesis-related parameters noninvasively during tumor progression. Typically, tumor tissue displays a high degree of heterogeneity that can be evaluated using pattern analysis (PA), which comprises shape and texture analysis. This work aims at implementing PA methods to study angiogenesis in a murine tumor model and testing its sensitivity with regard to detecting changes elicited by administration of a drug. Twelve balb/c-nude mice were injected subcutaneously with 10(6) C51 cells (colon carcinoma). A first group (N = 6) of animals was treated with dimethyloxalylglycine, a drug known to stabilize hypoxia-inducible-factor-α, which among other functions, is involved in angiogenesis. The second group (N = 6) was treated with saline. MRI experiments assessing tumor blood volume and permeability-maps (K(trans) ) were performed immediately before and 6 days after drug treatment. Data have been analyzed using standard histogram analysis and PA. Standard histogram analysis did not reveal any difference between the two groups, neither before nor after the treatment. In contrast, PA revealed significant differences between drug and placebo treated mice in the texture of the TBV and K(trans) maps after drug treatment, but not with regard to tumors shapes. The results indicated that in view of the heterogeneity of tumor tissue, standard histogram analysis appears insensitive in picking-up differences in response to treatment, while PA appears to be particularly sensitive to changes in texture.


Assuntos
Aminoácidos Dicarboxílicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Imageamento por Ressonância Magnética/métodos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Reconhecimento Automatizado de Padrão/métodos , Algoritmos , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/complicações , Aumento da Imagem/métodos , Interpretação de Imagem Assistida por Computador/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/complicações , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Resultado do Tratamento , Carga Tumoral
6.
Lab Invest ; 91(12): 1684-94, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21876537

RESUMO

Dimethyloxalylglycine (DMOG) is an inhibitor of prolyl-4-hydroxylase domain enzymes. Its potential value and mechanism of actions in preventing/treating gastrointestinal injury are, however, poorly understood. We, therefore, examined the effect of DMOG on influencing gut injury and repair using a variety of in vitro and in vivo models. We performed in vitro studies utilising pro-migratory (wounded monolayer) and proliferation (using DNA quantitation) assays of human stomach (AGS) and colonic (HT29) carcinoma cells. Time course studies examined changes in hypoxia-inducible factor (HIF) and vascular endothelial growth factor (VEGF) levels, a growth factor known to be regulated via HIF. In vivo studies utilised a rat gastric (indomethacin, 20 mg/kg and 3 h restraint) damage model. DMOG stimulated migration in a dose-dependent manner, increasing migration twofold when added at 25µM (P<0.01). Additive effects were seen when DMOG was added to cells in hypoxic conditions. DMOG stimulated proliferation dose dependently, increasing proliferation threefold when added at 70 µM (P<0.01). DMOG caused upregulation of both HIF and VEGF within 4 h of administration. Addition of VEGF neutralising antibody truncated migratory and proliferative activity of DMOG by about 70%. Both oral and subcutaneous administration of DMOG decreased gastric injury without influencing intragastric pH (50% reduction in injury when 1 ml gavaged at 0.57 mM, P < 0.01). Indomethacin reduced tissue HIF and VEGF levels but this was prevented if DMOG was present. In conclusion, DMOG stimulates the early phases of gut repair and VEGF-dependent processes appear relevant. Non-peptide factors such as this may be useful to stabilise or repair gut mucosa.


Assuntos
Aminoácidos Dicarboxílicos/uso terapêutico , Gastroenteropatias/tratamento farmacológico , Fator 1 Induzível por Hipóxia/metabolismo , Regeneração/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Aminoácidos Dicarboxílicos/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Células HT29 , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
7.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(3): 322-5, 2011 May.
Artigo em Chinês | MEDLINE | ID: mdl-21826991

RESUMO

OBJECTIVE: To investigate the effects of dimethyloxalyl glycine on hypoxic-ischemic brain damage in newborn rats. METHODS: Forty eight postnatal day 10 SD rats were divided into 3 groups, including sham surgery group, hypoxic-ischemic group and DMOG treated group. The brain tissues were collected at 4, 8, 24 and 72 hours after the hypoxic-ischemic treatment. The expressions of hypoxia inducible factor-1alpha (HIF-1alfa) protein and anti apoptoticprotein cleaved caspase 3 (CC3) were detected by immunohistochemistry. The apoptotic cells were detected by TUNEL staining. RESULTS: The expression level of HIF-1alpha was significantly higher in DMOG treated group than in hypoxic-ischemic group. While the expression level of CC3 was lower and the number of tunel positive cells was fewer in DMOG treated group than that in hypoxic-ischemic group. CONCLUSION: Dimethyloxalyl glycine may play a neuro-protective role in hypoxic-ischemic brain damage in newborn rats by stabilizing HIF-1alpha.


Assuntos
Aminoácidos Dicarboxílicos/uso terapêutico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Animais Recém-Nascidos , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
8.
Shock ; 36(3): 295-302, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21844787

RESUMO

Localized tissue hypoxia is a feature of infection and inflammation, resulting in the upregulation of the transcription factors hypoxia-inducible factor 1α and nuclear factor κB (NF-κB) via inhibition of oxygen sensing hydroxylase enzymes. Previous studies have demonstrated a beneficial role for the hydroxylase inhibitor dimethyloxallyl glycine (DMOG) in inflammatory conditions, including experimental colitis, by regulating the activity of hypoxia-inducible factor 1 and NF-κB. We have demonstrated in vivo that pretreatment with DMOG attenuates systemic LPS-induced activation of the NF-κB pathway. Furthermore, mice treated with DMOG had significantly increased survival in LPS-induced shock. Conversely, in models of polymicrobial sepsis, DMOG exacerbates disease severity. Dimethyloxallyl glycine treatment of mice promotes M2 polarization in macrophages within the peritoneal cavity, resulting in the downregulation of proinflammatory cytokines such as TNF-α. In addition, in vivo DMOG treatment upregulates IL-10 expression, specifically in the peritoneal B1 cell population. This study demonstrates cell type-specific roles for hydroxylase inhibition in vivo and provides insight into the mechanism underlying the protection conveyed by DMOG in models of endotoxic shock.


Assuntos
Aminoácidos Dicarboxílicos/uso terapêutico , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Choque Séptico/tratamento farmacológico , Animais , Citometria de Fluxo , Immunoblotting , Interleucina-10/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Oxigenases de Função Mista/antagonistas & inibidores , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase , Receptores de Interleucina-10/sangue , Sepse/tratamento farmacológico , Choque Séptico/induzido quimicamente
9.
J Immunol ; 186(7): 4367-74, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21357264

RESUMO

Intestinal ischemia/reperfusion injury (IR) is characterized by intermittent loss of perfusion to the gut, resulting in dramatic increases in morbidity and mortality. Based on previous studies indicating an anti-inflammatory role for hypoxia-inducible factor (HIF)-1-elicited enhancement of extracellular adenosine production via ecto-5'-nucleotidase (CD73) and signaling through the A2B adenosine receptor (A2BAR), we targeted HIF-1 during IR using pharmacological or genetic approaches. Initial studies with pharmacological HIF activation indicated attenuation of intestinal injury with dimethyloxallyl glycine (DMOG) treatment during murine IR. Although DMOG treatment was associated with induction of CD73 transcript and protein, DMOG protection was abolished in cd73(-/-) mice. Similarly, DMOG treatment enhanced A2BAR transcript and protein levels, whereas DMOG protection was abolished in A2BAR(-/-) mice. Finally, studies of mice with conditional HIF-1α deletion in intestinal epithelia or pharmacological inhibition of HIF-1 with 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin revealed enhanced tissue injury during IR. These studies indicated a tissue-protective role of HIF-dependent enhancement of intestinal adenosine generation and signaling during intestinal IR.


Assuntos
5'-Nucleotidase/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/uso terapêutico , Receptor A2B de Adenosina/fisiologia , Traumatismo por Reperfusão/prevenção & controle , 5'-Nucleotidase/deficiência , 5'-Nucleotidase/genética , Aminoácidos Dicarboxílicos/metabolismo , Aminoácidos Dicarboxílicos/uso terapêutico , Animais , Colite/enzimologia , Colite/fisiopatologia , Colite/prevenção & controle , Glicina/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/biossíntese , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
10.
Nat Rev Drug Discov ; 8(2): 139-52, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19165233

RESUMO

Cells in the human body need oxygen to function and survive, and severe deprivation of oxygen, as occurs in ischaemic heart disease and stroke, is a major cause of mortality. Nevertheless, other organisms, such as the fossorial mole rat or diving seals, have acquired the ability to survive in conditions of limited oxygen supply. Hypoxia tolerance also allows the heart to survive chronic oxygen shortage, and ischaemic preconditioning protects tissues against lethal hypoxia. The recent discovery of a new family of oxygen sensors--including prolyl hydroxylase domain-containing proteins 1-3 (PHD1-3)--has yielded exciting novel insights into how cells sense oxygen and keep oxygen supply and consumption in balance. Advances in understanding of the role of these oxygen sensors in hypoxia tolerance, ischaemic preconditioning and inflammation are creating new opportunities for pharmacological interventions for ischaemic and inflammatory diseases.


Assuntos
Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Hipóxia/prevenção & controle , Inflamação/tratamento farmacológico , Isquemia/tratamento farmacológico , Precondicionamento Isquêmico/métodos , Consumo de Oxigênio/efeitos dos fármacos , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Aminoácidos Dicarboxílicos/farmacologia , Aminoácidos Dicarboxílicos/uso terapêutico , Indutores da Angiogênese/uso terapêutico , Animais , Humanos , Hipóxia/metabolismo , Inflamação/metabolismo , Isquemia/metabolismo , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Consumo de Oxigênio/fisiologia
11.
Neurobiol Dis ; 31(3): 433-41, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18602008

RESUMO

Hypoxia-inducible factor-1alpha (HIF-1alpha) has been considered as a regulator of both prosurvival and prodeath pathways in the nervous system. The present study was designed to elucidate the role of HIF-1alpha in neonatal hypoxic-ischemic (HI) brain injury. Rice-Vannucci model of neonatal hypoxic-ischemic brain injury was used in seven-day-old rats, by subjecting unilateral carotid artery ligation followed by 2 h of hypoxia (8% O2 at 37 degrees C). HIF-1alpha activity was inhibited by 2-methoxyestradiol (2ME2) and enhanced by dimethyloxalylglycine (DMOG). Results showed that 2ME2 exhibited dose-dependent neuroprotection by decreasing infarct volume and reducing brain edema at 48 h post HI. The neuroprotection was lost when 2ME2 was administered 3 h post HI. HIF-1alpha upregulation by DMOG increased the permeability of the BBB and brain edema compared with HI group. 2ME2 attenuated the increase in HIF-1alpha and VEGF 24 h after HI. 2ME2 also had a long-term effect of protecting against the loss of brain tissue. The study showed that the early inhibition of HIF-1alpha acutely after injury provided neuroprotection after neonatal hypoxia-ischemia which was associated with preservation of BBB integrity, attenuation of brain edema, and neuronal death.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , 2-Metoxiestradiol , Aminoácidos Dicarboxílicos/farmacologia , Aminoácidos Dicarboxílicos/uso terapêutico , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/fisiopatologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatologia , Infarto Encefálico/tratamento farmacológico , Infarto Encefálico/metabolismo , Infarto Encefálico/fisiopatologia , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Hipóxia-Isquemia Encefálica/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Moduladores de Tubulina/farmacologia , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Exp Neurol ; 184 Suppl 1: S30-6, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14597323

RESUMO

Metabotropic glutamate receptors (mGluRs) are G-protein-coupled excitatory amino acid (glutamate) receptors and are abundantly expressed in basal ganglia nuclei. We used behavioral, regional glucose uptake metabolic mapping, and FOS protein expression to examine the effects of stimulating striatal and subthalamic mGluRs in rats. Stimulation of striatal Group I mGluRs produced behavioral effects mediated by polysynaptic activation of subthalamic neurons. Stimulation of subthalamic Group II mGluRs produced similar effects. Excessive activity of subthalamic neurons is a key feature of parkinsonism. mGluR Group I or Group II antagonists may prove to be useful for symptomatic treatment of parkinsonism. Stimulation of Group III mGluRs produced behavioral effects in only 6-hydroxydopamine-lesioned animals. Regional glucose uptake metabolic mapping and FOS expression studies suggested that striatal dopamine denervation produced increased sensitivity of Group III mGluRs. Agents active at Group III mGluRs may also be useful for treatment of parkinsonism.


Assuntos
Agonistas de Aminoácidos Excitatórios/uso terapêutico , Metoxi-Hidroxifenilglicol/análogos & derivados , Doença de Parkinson/tratamento farmacológico , Receptores de Glutamato Metabotrópico/agonistas , Aminoácidos Dicarboxílicos/farmacologia , Aminoácidos Dicarboxílicos/uso terapêutico , Animais , Autorradiografia , Gânglios da Base/anatomia & histologia , Gânglios da Base/efeitos dos fármacos , Gânglios da Base/metabolismo , Comportamento Animal , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glucose/metabolismo , Imuno-Histoquímica , Metoxi-Hidroxifenilglicol/farmacologia , Metoxi-Hidroxifenilglicol/uso terapêutico , Oxidopamina , Doença de Parkinson/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Receptores de Glutamato Metabotrópico/antagonistas & inibidores
13.
Br J Pharmacol ; 120(6): 1015-20, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9134211

RESUMO

1. We have previously shown that chronic antagonism of group I metabotropic glutamate receptors (mGluRs), in the brain, attenuates the precipitated morphine withdrawal syndrome in rats. In the present investigation we assessed the effects of chronic antagonism of group II and III mGluRs on the severity of withdrawal symptoms in rats treated chronically with subcutaneous (s.c.) morphine. 2. Concurrently with s.c. morphine we infused intracerebroventricularly (i.c.v.) one of a series of phenylglycine derivatives selective for specific mGluR subtypes. Group II mGluRs (mGluR2,3), which are negatively coupled to adenosine 3':5'-cyclic monophosphate (cyclic AMP) production, were selectively antagonized with 2s, 1's, 2's-2-methyl-2-(2'-carboxycyclopropyl) glycine (MCCG). Group III mGluRs (mGluR4,6,7 and 8), which are also negatively linked to cyclic AMP production, were selectively antagonized with alpha-methyl-L-amino-4-phosphonobutanoate (MAP4). The effects of MCCG and MAP4 were compared with alpha-methyl-4-carboxyphenylglycine (MCPG), which non-selectively antagonizes group II mGluRs, as well as group I mGluRs (mGluR1,5) which are positively coupled to phosphatidylinositol (PI) hydrolysis. 3. Chronic i.c.v. administration of both MCCG and MAP4 significantly decreased the time spent in withdrawal, MCPG and MCCG reduced the frequency of jumps and wet dog shakes and attenuated the severity of agitation. 4. Acute i.c.v. injection of mGluR antagonists just before the precipitation of withdrawal failed to decrease the severity of abstinence symptoms. Rather, acute i.c.v. injection of MCCG significantly increased the time spent in withdrawal. 5. Our results suggest that the development of opioid dependence is affected by mGluR-mediated PI hydrolysis and mGluR-regulated cyclic AMP production.


Assuntos
Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Dependência de Morfina/tratamento farmacológico , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Aminoácidos Dicarboxílicos/uso terapêutico , Aminobutiratos/uso terapêutico , Animais , Benzoatos/uso terapêutico , Glicina/análogos & derivados , Glicina/uso terapêutico , Injeções Intraventriculares , Masculino , Dependência de Morfina/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA