Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Ann Clin Transl Neurol ; 8(7): 1508-1514, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34053190

RESUMO

Schwannomas are benign neoplasms that can cause gain- and loss-of-function neurological phenotypes, including severe, intractable pain. To investigate the molecular mechanisms underlying schwannoma-associated pain we compared the RNA sequencing profile of painful and non-painful schwannomas from NF2 patients. Distinct segregation of painful and non-painful tumors by gene expression patterns was observed. Differential expression analysis showed the upregulation of fibroblast growth factor 7 (FGF7) in painful schwannomas. Behavioral support for this finding was observed using a xenograft human NF2-schwannoma model in nude mice. In this model, over-expression of FGF7 in intra-sciatically implanted NF2 tumor cells generated pain behavior compared with controls.


Assuntos
Fator 7 de Crescimento de Fibroblastos/genética , Neurilemoma/genética , Neurofibromatose 2/genética , Dor/genética , Análise de Sequência de RNA/métodos , Transcriptoma/genética , Animais , Linhagem Celular Tumoral , Feminino , Fator 7 de Crescimento de Fibroblastos/biossíntese , Humanos , Masculino , Camundongos , Camundongos Nus , Neurilemoma/metabolismo , Neurilemoma/patologia , Neurofibromatose 2/metabolismo , Neurofibromatose 2/patologia , Dor/metabolismo , Dor/patologia , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
Dalton Trans ; 49(1): 17-22, 2020 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-31799569

RESUMO

The Sr0.95Sm0.0125Dy0.0125□0.025Ti0.90Nb0.10O3±Î´/ZrO2 composite was directly prepared through spark plasma sintering. This approach limited the grain growth and facilitated the achievement of a narrow grain size distribution due to fast sintering and ZrO2 effects. Thermal conductivity declined to 1.68 W m-1 K-1, which is the lowest among the reported values for micro-polycrystalline SrTiO3-based structures.

3.
Pain ; 160(11): 2524-2534, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31246732

RESUMO

A recently defined structure, the rostromedial tegmental nucleus (RMTg; aka tail of the ventral tegmental area [VTA]), has been proposed as an inhibitory control center for dopaminergic activity of the VTA. This region is composed of GABAergic cells that send afferent projections to the ventral midbrain and synapse onto dopaminergic cells in the VTA and substantia nigra. These cells exhibit µ-opioid receptor immunoreactivity, and in vivo, ex vivo, and optogenetic/electrophysiological approaches demonstrate that morphine excites dopamine neurons by targeting receptors on GABAergic neurons localized in the RMTg. This suggests that the RMTg may be a key modulator of opioid effects and a major brake regulating VTA dopamine systems. However, no study has directly manipulated RMTg GABAergic neurons in vivo and assessed the effect on nociception or opioid analgesia. In this study, multiplexing of GABAergic neurons in the RMTg was achieved using stimulatory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) and inhibitory kappa-opioid receptor DREADDs (KORD). Our data show that locally infused RMTg morphine or selective RMTg GABAergic neuron inhibition produces 87% of the maximal antinociceptive effect of systemic morphine, and RMTg GABAergic neurons modulate dopamine release in the nucleus accumbens. In addition, chemoactivation of VTA dopamine neurons significantly reduced pain behaviors both in resting and facilitated pain states and reduced by 75% the dose of systemic morphine required to produce maximal antinociception. These results provide compelling evidence that RMTg GABAergic neurons are involved in processing of nociceptive information and are important mediators of opioid analgesia.


Assuntos
Analgésicos Opioides/farmacologia , Vias Neurais/efeitos dos fármacos , Tegmento Mesencefálico/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Camundongos Transgênicos , Morfina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores Opioides/efeitos dos fármacos , Tegmento Mesencefálico/citologia , Ácido gama-Aminobutírico/farmacologia
4.
Neuro Oncol ; 21(7): 854-866, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-30977509

RESUMO

BACKGROUND: We evaluated apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) as a schwannoma tumor suppressor and explored its utilization in a schwannoma gene therapy strategy that may be translated to clinical use. METHODS: ASC protein expression and mRNA level were assessed in human schwannoma by immunohistochemistry and quantitative PCR, respectively. Methylation- specific PCR was used to assess ASC promoter methylation. The effect of ASC overexpression in schwannoma cells was evaluated through ATP-based viability, lactate dehydrogenase release, and apoptosis staining. Western blotting and colorimetric assay were used to test the effect of ASC overexpression on endogenous pro-apoptotic pathways. Bioluminescence imaging, behavioral testing, and immunohistochemistry in human xenograft and murine allograft schwannoma models were used to examine the efficacy and toxicity of intratumoral injection of adeno-associated virus (AAV) vector encoding ASC. RESULTS: ASC expression was suppressed via promoter methylation in over 80% of the human schwannomas tested. ASC overexpression in schwannoma cells results in cell death and is associated with activation of endogenous caspase-9, caspase-3, and upregulation of BH3 interacting-domain death agonist. In a human xenograft schwannoma model, AAV1-mediated ASC delivery reduced tumor growth and resolved tumor-associated pain without detectable toxicity, and tumor control was associated with reduced Ki67 mitotic index and increased tumor-cell apoptosis. Efficacy of this schwannoma gene therapy strategy was confirmed in a murine schwannoma model. CONCLUSION: We have identified ASC as a putative schwannoma tumor suppressor with high potential clinical utility for schwannoma gene therapy and generated a vector that treats schwannomas via a novel mechanism that does not overlap with current treatments.


Assuntos
Apoptose , Biomarcadores Tumorais/genética , Proteínas Adaptadoras de Sinalização CARD/administração & dosagem , Dor do Câncer/prevenção & controle , Terapia Genética , Neurilemoma/terapia , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Dor do Câncer/etiologia , Proliferação de Células , Metilação de DNA , Dependovirus/genética , Humanos , Masculino , Camundongos , Neurilemoma/genética , Neurilemoma/patologia , Prognóstico , Regiões Promotoras Genéticas , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa