Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 199: 106575, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38914170

RESUMO

CT1812 is a novel, brain penetrant small molecule modulator of the sigma-2 receptor (S2R) that is currently in clinical development for the treatment of Alzheimer's disease (AD). Preclinical and early clinical data show that, through S2R, CT1812 selectively prevents and displaces binding of amyloid beta (Aß) oligomers from neuronal synapses and improves cognitive function in animal models of AD. SHINE is an ongoing phase 2 randomized, double-blind, placebo-controlled clinical trial (COG0201) in participants with mild to moderate AD, designed to assess the safety and efficacy of 6 months of CT1812 treatment. To elucidate the mechanism of action in AD patients and pharmacodynamic biomarkers of CT1812, the present study reports exploratory cerebrospinal fluid (CSF) biomarker data from 18 participants in an interim analysis of the first set of patients in SHINE (part A). Untargeted mass spectrometry-based discovery proteomics detects >2000 proteins in patient CSF and has documented utility in accelerating the identification of novel AD biomarkers reflective of diverse pathophysiologies beyond amyloid and tau, and enabling identification of pharmacodynamic biomarkers in longitudinal interventional trials. We leveraged this technique to analyze CSF samples taken at baseline and after 6 months of CT1812 treatment. Proteome-wide protein levels were detected using tandem mass tag-mass spectrometry (TMT-MS), change from baseline was calculated for each participant, and differential abundance analysis by treatment group was performed. This analysis revealed a set of proteins significantly impacted by CT1812, including pathway engagement biomarkers (i.e., biomarkers tied to S2R biology) and disease modification biomarkers (i.e., biomarkers with altered levels in AD vs. healthy control CSF but normalized by CT1812, and biomarkers correlated with favorable trends in ADAS-Cog11 scores). Brain network mapping, Gene Ontology, and pathway analyses revealed an impact of CT1812 on synapses, lipoprotein and amyloid beta biology, and neuroinflammation. Collectively, the findings highlight the utility of this method in pharmacodynamic biomarker identification and providing mechanistic insights for CT1812, which may facilitate the clinical development of CT1812 and enable appropriate pre-specification of biomarkers in upcoming clinical trials of CT1812.

2.
Neurobiol Dis ; 181: 106128, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37075948

RESUMO

RGS14 is a complex multifunctional scaffolding protein that is highly enriched within pyramidal cells (PCs) of hippocampal area CA2. In these neurons, RGS14 suppresses glutamate-induced calcium influx and related G protein and ERK signaling in dendritic spines to restrain postsynaptic signaling and plasticity. Previous findings show that, unlike PCs of hippocampal areas CA1 and CA3, CA2 PCs are resistant to a number of neurological insults, including degeneration caused by temporal lobe epilepsy (TLE). While RGS14 is protective against peripheral injury, similar roles for RGS14 during pathological injury in hippocampus remain unexplored. Recent studies showed that area CA2 modulates hippocampal excitability, generates epileptiform activity and promotes hippocampal pathology in animal models and patients with TLE. Because RGS14 suppresses CA2 excitability and signaling, we hypothesized that RGS14 would moderate seizure behavior and early hippocampal pathology following seizure activity, possibly affording protection to CA2 PCs. Using kainic acid (KA) to induce status epilepticus (KA-SE) in mice, we show that the loss of RGS14 (RGS14 KO) accelerated onset of limbic motor seizures and mortality compared to wild type (WT) mice, and that KA-SE upregulated RGS14 protein expression in CA2 and CA1 PCs of WT. Our proteomics data show that the loss of RGS14 impacted the expression of a number of proteins at baseline and after KA-SE, many of which associated unexpectedly with mitochondrial function and oxidative stress. RGS14 was shown to localize to the mitochondria in CA2 PCs of mice and reduce mitochondrial respiration in vitro. As a readout of oxidative stress, we found that RGS14 KO dramatically increased 3- nitrotyrosine levels in CA2 PCs, which was greatly exacerbated following KA-SE and correlated with a lack of superoxide dismutase 2 (SOD2) induction. Assessing for hallmarks of seizure pathology in RGS14 KO, we unexpectedly found no differences in neuronal injury in CA2 PCs. However, we observed a striking and surprising lack of microgliosis in CA1 and CA2 of RGS14 KO compared to WT. Together, our data demonstrate a newly appreciated role for RGS14 in limiting intense seizure activity and pathology in hippocampus. Our findings are consistent with a model where RGS14 limits seizure onset and mortality and, after seizure, is upregulated to support mitochondrial function, prevent oxidative stress in CA2 PCs, and promote microglial activation in hippocampus.


Assuntos
Epilepsia do Lobo Temporal , Proteínas RGS , Estado Epiléptico , Animais , Camundongos , Hipocampo/metabolismo , Convulsões , Células Piramidais/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Estresse Oxidativo , Ácido Caínico/toxicidade , Proteínas RGS/efeitos adversos , Proteínas RGS/metabolismo
3.
Mol Psychiatry ; 22(1): 13-23, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27872486

RESUMO

Preclinical changes that precede the onset of symptoms and eventual diagnosis of Alzheimer's disease (AD) are a target for potential preventive interventions. A large body of evidence suggests that inflammation is closely associated with AD pathogenesis and may be a promising target pathway for such interventions. However, little is known about the association between systemic inflammation and preclinical AD pathophysiology. We first examined whether the acute-phase protein, alpha-2 macroglobulin (A2M), a major component of the innate immune system, was associated with cerebrospinal fluid (CSF) markers of neuronal injury in preclinical AD and risk of incident AD in the predictors of cognitive decline among normal individuals (BIOCARD) cohort. We find that A2M concentration in blood is significantly associated with CSF concentrations of the neuronal injury markers, tau and phosphorylated tau, and that higher baseline serum A2M concentration is associated with an almost threefold greater risk of progression to clinical symptoms of AD in men. These findings were replicated in the Alzheimer's Disease Neuroimaging (ADNI) study. Then, utilizing a systems level approach combining large multi-tissue gene expression datasets with mass spectrometry-based proteomic analyses of brain tissue, we identified an A2M gene network that includes regulator of calcineurin (RCAN1), an inhibitor of calcineurin, a well-characterized tau phosphatase. A2M gene and protein expression in the brain were significantly associated with gene and protein expression levels of calcineurin. Collectively these novel findings suggest that A2M is associated with preclinical AD, reflects early neuronal injury in the disease course and may be responsive to tau phosphorylation in the brain through the RCAN1-calcineurin pathway.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Musculares/metabolismo , alfa-Macroglobulinas/metabolismo , Idoso , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Biomarcadores/líquido cefalorraquidiano , Encéfalo/metabolismo , Calcineurina , Cognição/fisiologia , Transtornos Cognitivos/metabolismo , Estudos de Coortes , Proteínas de Ligação a DNA , Progressão da Doença , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Imunidade Inata , Inflamação/líquido cefalorraquidiano , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Neuroimagem , Neurônios , Fosforilação , Proteômica , alfa-Macroglobulinas/análise , Proteínas tau/metabolismo
4.
Nat Biotechnol ; 19(1): 35-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11135549

RESUMO

Research studies suggest that tumor-related angiogenesis contributes to the phenotype of malignant gliomas. We assessed the effect of local delivery of the angiogenesis inhibitor endostatin on human glioma cell line (U-87MG) xenografts. Baby hamster kidney (BHK) cells were stably transfected with a human endostatin (hES) expression vector and were encapsulated in alginate-poly L-lysine (PLL) microcapsules for long-term delivery of hES. The release of biologically active endostatin was confirmed using assays of bovine capillary endothelial (BCE) proliferation and of tube formation. Human endostatin released from the microcapsules brought about a 67. 2% inhibition of BCE proliferation. Furthermore, secreted hES was able to inhibit tube formation in KDR/PAE cells (porcine aortic endothelial cells stably transfected with KDR, a tyrosine kinase) treated with conditioned U-87MG medium. A single local injection of encapsulated endostatin-secreting cells in a nude mouse model resulted in a 72.3% reduction in subcutaneous U87 xenografts' weight 21 days post treatment. This inhibition was achieved by only 150.8 ng/ml human endostatin secreted from 2 x 10(5) encapsulated cells. Encapsulated endostatin-secreting cells are effective for the treatment of human glioblastoma xenografts. Continuous local delivery of endostatin may offer an effective therapeutic approach to the treatment of a variety of tumor types.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neoplasias Encefálicas/terapia , Colágeno/administração & dosagem , Colágeno/genética , Glioma/terapia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Alginatos , Inibidores da Angiogênese/uso terapêutico , Inibidores da Angiogênese/toxicidade , Animais , Materiais Biocompatíveis , Capilares , Cápsulas , Bovinos , Transplante de Células , Células Cultivadas , Colágeno/uso terapêutico , Cricetinae , Endostatinas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Fragmentos de Peptídeos/uso terapêutico , Polilisina/análogos & derivados , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Suínos , Transfecção , Transplante Heterólogo
5.
Cancer Res ; 60(17): 4926-31, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10987308

RESUMO

The up-regulation of cyclooxygenase 2 (COX-2) expression is a frequent occurrence in a variety of different tumors. In this study, COX-2 protein expression was investigated in 50 glioma and 3 normal brain specimens by immunohistochemistry. Expression of COX-2 protein was observed in all normal brain and glioma specimens by immunohistochemistry, regardless of histological grade. The immunoreactive score was significantly higher in high-grade glioma than low-grade glioma and normal brain specimens. For a subset of these tumors (nine gliomas and three normal brain), Western blot analysis was also performed. COX-2 protein was detected in all specimens by Western blot analysis. The effect of the specific COX-2 inhibitor NS-398 on monolayer cell cultures and three-dimensional glioma spheroids was investigated using U-87MG and U-251MG human glioblastoma cell lines. The proliferation rate was assessed in monolayer cultures. In addition, a growth assay, a migration assay, an apoptosis assay, and a tumor invasion assay were performed in a three-dimensional spheroid culture system. NS-398 was able to reduce the proliferation of monolayer cell cultures, as well as the growth of spheroids and tumor cell migration, in a dose-dependent manner. There was also a moderate increase in the number of apoptotic cells in the treated spheroids. NS-398 did not have an inhibitory effect on tumor invasion in the coculture spheroid system. Our study provides evidence that COX-2 is up-regulated in the majority of high-grade gliomas and that a potential role of COX-2 inhibitors as an adjuvant therapy for brain tumors may exist.


Assuntos
Astrocitoma/enzimologia , Neoplasias Encefálicas/enzimologia , Inibidores de Ciclo-Oxigenase/farmacologia , Glioblastoma/enzimologia , Isoenzimas/biossíntese , Nitrobenzenos/farmacologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Sulfonamidas/farmacologia , Adulto , Animais , Apoptose/efeitos dos fármacos , Astrocitoma/tratamento farmacológico , Astrocitoma/patologia , Encéfalo/enzimologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Técnicas de Cocultura , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Inibidores do Crescimento/farmacologia , Humanos , Isoenzimas/antagonistas & inibidores , Masculino , Proteínas de Membrana , Pessoa de Meia-Idade , Invasividade Neoplásica , Ratos , Ratos Sprague-Dawley , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Células Tumorais Cultivadas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...