Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Ther Oncolytics ; 26: 49-62, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35784402

RESUMO

Genetically engineered neural stem cells (NSCs) are a promising therapy for the highly aggressive brain cancer glioblastoma (GBM); however, treatment durability remains a major challenge. We sought to define the events that contribute to dynamic adaptation of GBM during treatment with human skin-derived induced NSCs releasing the pro-apoptotic agent TRAIL (iNSC-TRAIL) and develop strategies that convert initial tumor kill into sustained GBM suppression. In vivo and ex vivo analysis before, during, and after treatment revealed significant shifts in tumor transcriptome and spatial distribution as the tumors adapted to treatment. To address this, we designed iNSC delivery strategies that increased spatiotemporal TRAIL coverage and significantly decreased GBM volume throughout the brain, reducing tumor burden 100-fold as quantified in live ex vivo brain slices. The varying impact of different strategies on treatment durability and median survival of both solid and invasive tumors provides important guidance for optimizing iNSC therapy.

2.
Genes Dev ; 34(7-8): 526-543, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32079652

RESUMO

MDM2 and MDMX, negative regulators of the tumor suppressor p53, can work separately and as a heteromeric complex to restrain p53's functions. MDM2 also has pro-oncogenic roles in cells, tissues, and animals that are independent of p53. There is less information available about p53-independent roles of MDMX or the MDM2-MDMX complex. We found that MDM2 and MDMX facilitate ferroptosis in cells with or without p53. Using small molecules, RNA interference reagents, and mutant forms of MDMX, we found that MDM2 and MDMX, likely working in part as a complex, normally facilitate ferroptotic death. We observed that MDM2 and MDMX alter the lipid profile of cells to favor ferroptosis. Inhibition of MDM2 or MDMX leads to increased levels of FSP1 protein and a consequent increase in the levels of coenzyme Q10, an endogenous lipophilic antioxidant. This suggests that MDM2 and MDMX normally prevent cells from mounting an adequate defense against lipid peroxidation and thereby promote ferroptosis. Moreover, we found that PPARα activity is essential for MDM2 and MDMX to promote ferroptosis, suggesting that the MDM2-MDMX complex regulates lipids through altering PPARα activity. These findings reveal the complexity of cellular responses to MDM2 and MDMX and suggest that MDM2-MDMX inhibition might be useful for preventing degenerative diseases involving ferroptosis. Furthermore, they suggest that MDM2/MDMX amplification may predict sensitivity of some cancers to ferroptosis inducers.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ferroptose/genética , Metabolismo dos Lipídeos/genética , PPAR alfa/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Proteínas de Ciclo Celular/genética , Glioblastoma/fisiopatologia , Células HCT116 , Humanos , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2/genética , Interferência de RNA , Ratos , Proteína Supressora de Tumor p53/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
3.
EBioMedicine ; 47: 33-43, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31466914

RESUMO

BACKGROUND: The dismal survival of glioblastoma (GBM) patients urgently calls for the development of new treatments. Chimeric antigen receptor T (CAR-T) cells are an attractive strategy, but preclinical and clinical studies in GBM have shown that heterogeneous expression of the antigens targeted so far causes tumor escape, highlighting the need for the identification of new targets. We explored if B7-H3 is a valuable target for CAR-T cells in GBM. METHODS: We compared mRNA expression of antigens in GBM using TCGA data, and validated B7-H3 expression by immunohistochemistry. We then tested the antitumor activity of B7-H3-redirected CAR-T cells against GBM cell lines and patient-derived GBM neurospheres in vitro and in xenograft murine models. FINDINGS: B7-H3 mRNA and protein are overexpressed in GBM relative to normal brain in all GBM subtypes. Of the 46 specimens analyzed by immunohistochemistry, 76% showed high B7-H3 expression, 22% had detectable, but low B7-H3 expression and 2% were negative, as was normal brain. All 20 patient-derived neurospheres showed ubiquitous B7-H3 expression. B7-H3-redirected CAR-T cells effectively targeted GBM cell lines and neurospheres in vitro and in vivo. No significant differences were found between CD28 and 4-1BB co-stimulation, although CD28-co-stimulated CAR-T cells released more inflammatory cytokines. INTERPRETATION: We demonstrated that B7-H3 is highly expressed in GBM specimens and neurospheres that contain putative cancer stem cells, and that B7-H3-redirected CAR-T cells can effectively control tumor growth. Therefore, B7-H3 represents a promising target in GBM. FUND: Alex's Lemonade Stand Foundation; Il Fondo di Gio Onlus; National Cancer Institute; Burroughs Wellcome Fund.


Assuntos
Antígenos B7/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Antígenos B7/genética , Biomarcadores , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioblastoma/imunologia , Glioblastoma/mortalidade , Glioblastoma/terapia , Humanos , Imunofenotipagem , Imunoterapia Adotiva , Camundongos , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Neuro Oncol ; 21(12): 1552-1564, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31420675

RESUMO

BACKGROUND: Tumor-homing tumoricidal neural stem cell (tNSC) therapy is a promising new strategy that recently entered human patient testing for glioblastoma (GBM). Developing strategies for tNSC therapy to overcome intratumoral heterogeneity, variable cancer cell invasiveness, and differential drug response of GBM will be essential for efficacious treatment response in the clinical setting. The aim of this study was to create novel hybrid tumor models and investigate the impact of GBM heterogeneity on tNSC therapies. METHODS: We used organotypic brain slice explants and distinct human GBM cell types to generate heterogeneous models ex vivo and in vivo. We then tested the efficacy of mono- and combination therapy with primary NSCs and fibroblast-derived human induced neural stem cells (iNSCs) engineered with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or enzyme-prodrug therapy. RESULTS: Optical imaging, molecular assays, and immunohistochemistry revealed that the hybrid models recapitulated key aspects of patient GBM, including heterogeneity in TRAIL sensitivity, proliferation, migration patterns, hypoxia, blood vessel structure, cancer stem cell populations, and immune infiltration. To explore the impact of heterogeneity on tNSC therapy, testing in multiple in vivo models showed that tNSC-TRAIL therapy potently inhibited tumor growth and significantly increased survival across all paradigms. Patterns of tumor recurrence varied with therapeutic (tNSC-TRAIL and/or tNSC-thymidine kinase), dose, and route of administration. CONCLUSIONS: These studies report new hybrid models that accurately capture key aspects of GBM heterogeneity which markedly impact treatment response while demonstrating the ability of tNSC mono- and combination therapy to overcome certain aspects of heterogeneity for robust tumor kill.


Assuntos
Glioblastoma/patologia , Glioblastoma/terapia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/citologia , Transplante de Células-Tronco/métodos , Animais , Apoptose , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Nus , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Hum Mol Genet ; 26(21): 4267-4277, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973132

RESUMO

Neuronal interleukin-34 (IL-34) promotes the expansion of microglia in the central nervous system-microglial activation and expansion are in turn implicated in the pathogenesis of Huntington's disease (HD). We thus examined whether the accumulation of an amyloidogenic exon-1 fragment of mutant huntingtin (mHTTx1) modulates the expression of IL-34 in dopaminergic neurons derived from a human embryonic stem cell line. We found that mHTTx1 aggregates induce IL-34 production selectively in post-mitotic neurons. Exposure of neurons to DNA damaging agents or the excitotoxin NMDA elicited similar results suggesting that IL-34 induction may be a general response to neuronal stress including the accumulation of misfolded mHTTx1. We further determined that knockdown or blocking the activity of IκB kinase beta (IKKß) prevented the aggregation of mHTTx1 and subsequent IL-34 production. While elevated IL-34 itself had no effect on the aggregation or the toxicity of mHTTx1 in neuronal culture, IL-34 expression in a rodent brain slice model with intact neuron-microglial networks exacerbated mHTTx1-induced degeneration of striatal medium-sized spiny neurons. Conversely, an inhibitor of the IL-34 receptor reduced microglial numbers and ameliorated mHTTx1-mediated neurodegeneration. Together, these findings uncover a novel function for IKKß/mHTTx1 interactions in regulating IL-34 production, and implicate a role for IL-34 in non-cell-autonomous, microglial-dependent neurodegeneration in HD.


Assuntos
Doença de Huntington/metabolismo , Doença de Huntington/patologia , Quinase I-kappa B/metabolismo , Interleucinas/metabolismo , Animais , Linhagem Celular , Corpo Estriado/metabolismo , Éxons , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Quinase I-kappa B/genética , Interleucinas/genética , Microglia/metabolismo , Microglia/patologia , Neurônios/metabolismo , Neurotoxinas/metabolismo , Cultura Primária de Células , Ratos
6.
Nat Commun ; 8(1): 623, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931805

RESUMO

The immense and growing repositories of transcriptional data may contain critical insights for developing new therapies. Current approaches to mining these data largely rely on binary classifications of disease vs. control, and are not able to incorporate measures of disease severity. We report an analytical approach to integrate ordinal clinical information with transcriptomics. We apply this method to public data for a large cohort of Huntington's disease patients and controls, identifying and prioritizing phenotype-associated genes. We verify the role of a high-ranked gene in dysregulation of sphingolipid metabolism in the disease and demonstrate that inhibiting the enzyme, sphingosine-1-phosphate lyase 1 (SPL), has neuroprotective effects in Huntington's disease models. Finally, we show that one consequence of inhibiting SPL is intracellular inhibition of histone deacetylases, thus linking our observations in sphingolipid metabolism to a well-characterized Huntington's disease pathway. Our approach is easily applied to any data with ordinal clinical measurements, and may deepen our understanding of disease processes.Identifying gene subsets affecting disease phenotypes from transcriptome data is challenge. Here, the authors develop a method that combines transcriptional data with disease ordinal clinical measurements to discover a sphingolipid metabolism regulator involving in Huntington's disease progression.


Assuntos
Aldeído Liases/genética , Doença de Huntington/genética , Células-Tronco Neurais/metabolismo , Aldeído Liases/antagonistas & inibidores , Aldeído Liases/metabolismo , Animais , Estudos de Casos e Controles , Estudos de Coortes , Humanos , Doença de Huntington/metabolismo , Doença de Huntington/fisiopatologia , Masculino , Camundongos , Neostriado/citologia , Fenótipo
7.
Oncotarget ; 7(43): 69173-69187, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27732951

RESUMO

Glioblastoma multiforme (GBM) carries a poor prognosis and continues to lack effective treatments. Glioblastoma stem cells (GSCs) drive tumor formation, invasion, and drug resistance and, as such, are the focus of studies to identify new therapies for disease control. Here, we identify the involvement of IKK and NF-κB signaling in the maintenance of GSCs. Inhibition of this pathway impairs self-renewal as analyzed in tumorsphere formation and GBM expansion as analyzed in brain slice culture. Interestingly, both the canonical and non-canonical branches of the NF-κB pathway are shown to contribute to this phenotype. One source of NF-κB activation in GBM involves the TGF-ß/TAK1 signaling axis. Together, our results demonstrate a role for the NF-κB pathway in GSCs and provide a mechanistic basis for its potential as a therapeutic target in glioblastoma.


Assuntos
Autorrenovação Celular , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Quinase I-kappa B/genética , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/genética , Interferência de RNA , Ratos , Esferoides Celulares/metabolismo , Técnicas de Cultura de Tecidos , Fator de Crescimento Transformador beta/metabolismo
8.
Sci Rep ; 6: 25626, 2016 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-27172999

RESUMO

We previously reported neuroprotective activity of the botanical anti-cancer drug candidate PBI-05204, a supercritical CO2 extract of Nerium oleander, in brain slice and in vivo models of ischemic stroke. We showed that one component of this neuroprotective activity is mediated through its principal cardiac glycoside constituent, oleandrin, via induction of the potent neurotrophic factor brain-derived neurotrophic factor (BDNF). However, we also noted that the concentration-relation for PBI-05204 in the brain slice oxygen-glucose deprivation (OGD) model is considerably broader than that for oleandrin as a single agent. We thus surmised that PBI-05204 contains an additional neuroprotective component(s), distinct from oleandrin. We report here that neuroprotective activity is also provided by the triterpenoid constituents of PBI-05204, notably oleanolic acid. We demonstrate that a sub-fraction of PBI-05204 (Fraction 0-4) containing oleanolic and other triterpenoids, but without cardiac glycosides, induces the expression of cellular antioxidant gene transcription programs regulated through antioxidant transcriptional response elements (AREs). Finally, we show that Fraction 0-4 provides broad neuroprotection in organotypic brain slice models for neurodegeneration driven by amyloid precursor protein (APP) and tau implicated in Alzheimer's disease and frontotemporal dementias, respectively, in addition to ischemic injury modeled by OGD.


Assuntos
Antineoplásicos/farmacologia , Encéfalo/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Doenças Neurodegenerativas/tratamento farmacológico , Extratos Vegetais/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Antineoplásicos/química , Encéfalo/metabolismo , Encéfalo/patologia , Fracionamento Químico/métodos , Modelos Animais de Doenças , Feminino , Glucose/metabolismo , Humanos , Masculino , Nerium/química , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Ácido Oleanólico/química , Ácido Oleanólico/farmacologia , Técnicas de Cultura de Órgãos , Oxigênio/metabolismo , Ratos Sprague-Dawley
9.
Proc Natl Acad Sci U S A ; 112(17): E2245-52, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25848045

RESUMO

Protein disulfide isomerase (PDI) is a chaperone protein in the endoplasmic reticulum that is up-regulated in mouse models of, and brains of patients with, neurodegenerative diseases involving protein misfolding. PDI's role in these diseases, however, is not fully understood. Here, we report the discovery of a reversible, neuroprotective lead optimized compound (LOC)14, that acts as a modulator of PDI. LOC14 was identified using a high-throughput screen of ∼10,000 lead-optimized compounds for potent rescue of viability of PC12 cells expressing mutant huntingtin protein, followed by an evaluation of compounds on PDI reductase activity in an in vitro screen. Isothermal titration calorimetry and fluorescence experiments revealed that binding to PDI was reversible with a Kd of 62 nM, suggesting LOC14 to be the most potent PDI inhibitor reported to date. Using 2D heteronuclear single quantum correlation NMR experiments, we were able to map the binding site of LOC14 as being adjacent to the active site and to observe that binding of LOC14 forces PDI to adopt an oxidized conformation. Furthermore, we found that LOC14-induced oxidation of PDI has a neuroprotective effect not only in cell culture, but also in corticostriatal brain slice cultures. LOC14 exhibited high stability in mouse liver microsomes and blood plasma, low intrinsic microsome clearance, and low plasma-protein binding. These results suggest that LOC14 is a promising lead compound to evaluate the potential therapeutic effects of modulating PDI in animal models of disease.


Assuntos
Corpo Estriado/enzimologia , Inibidores Enzimáticos , Microssomos Hepáticos/enzimologia , Fármacos Neuroprotetores , Isomerases de Dissulfetos de Proteínas , Animais , Domínio Catalítico , Corpo Estriado/citologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Proteína Huntingtina , Camundongos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Ressonância Magnética Nuclear Biomolecular , Oxirredução/efeitos dos fármacos , Células PC12 , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/química , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Ratos
10.
J Am Chem Soc ; 136(12): 4551-6, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24592866

RESUMO

Ferrostatin-1 (Fer-1) inhibits ferroptosis, a form of regulated, oxidative, nonapoptotic cell death. We found that Fer-1 inhibited cell death in cellular models of Huntington's disease (HD), periventricular leukomalacia (PVL), and kidney dysfunction; Fer-1 inhibited lipid peroxidation, but not mitochondrial reactive oxygen species formation or lysosomal membrane permeability. We developed a mechanistic model to explain the activity of Fer-1, which guided the development of ferrostatins with improved properties. These studies suggest numerous therapeutic uses for ferrostatins, and that lipid peroxidation mediates diverse disease phenotypes.


Assuntos
Cicloexilaminas/farmacologia , Doença de Huntington/tratamento farmacológico , Nefropatias/tratamento farmacológico , Leucomalácia Periventricular/tratamento farmacológico , Lipídeos de Membrana/metabolismo , Fenilenodiaminas/farmacologia , Morte Celular/efeitos dos fármacos , Cicloexilaminas/uso terapêutico , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Leucomalácia Periventricular/metabolismo , Leucomalácia Periventricular/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Fenilenodiaminas/uso terapêutico
11.
J Neurosci ; 34(3): 963-8, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24431454

RESUMO

We have previously shown that the botanical drug candidate PBI-05204, a supercritical CO2 extract of Nerium oleander, provides neuroprotection in both in vitro and in vivo brain slice-based models for focal ischemia (Dunn et al., 2011). Intriguingly, plasma levels of the neurotrophin BDNF were increased in patients treated with PBI-05204 in a phase I clinical trial (Bidyasar et al., 2009). We thus tested the hypothesis that neuroprotection provided by PBI-05204 to rat brain slices damaged by oxygen-glucose deprivation (OGD) is mediated by BDNF. We found, in fact, that exogenous BDNF protein itself is sufficient to protect brain slices against OGD, whereas downstream activation of TrkB receptors for BDNF is necessary for neuroprotection provided by PBI-05204, using three independent methods. Finally, we provide evidence that oleandrin, the principal cardiac glycoside component of PBI-05204, can quantitatively account for regulation of BDNF at both the protein and transcriptional levels. Together, these findings support further investigation of cardiac glycosides in providing neuroprotection in the context of ischemic stroke.


Assuntos
Antioxidantes/fisiologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Cardenolídeos/farmacologia , Glucose/deficiência , Fármacos Neuroprotetores/farmacologia , Consumo de Oxigênio/fisiologia , Oxigênio/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Masculino , Nerium , Técnicas de Cultura de Órgãos , Extratos Vegetais/farmacologia , Ratos , Ratos Sprague-Dawley
12.
J Neurochem ; 119(4): 805-14, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21950737

RESUMO

The principal active constituent of the botanical drug candidate PBI-05204, a supercritical CO(2) extract of Nerium oleander, is the cardiac glycoside oleandrin. PBI-05204 shows potent anticancer activity and is currently in phase I clinical trial as a treatment for patients with solid tumors. We have previously shown that neriifolin, which is structurally related to oleandrin, provides robust neuroprotection in brain slice and whole animal models of ischemic injury. However, neriifolin itself is not a suitable drug development candidate and the FDA-approved cardiac glycoside digoxin does not cross the blood-brain barrier. We report here that both oleandrin as well as the full PBI-05204 extract can also provide significant neuroprotection to neural tissues damaged by oxygen and glucose deprivation as occurs in ischemic stroke. Critically, we show that the neuroprotective activity of PBI-05204 is maintained for several hours of delay of administration after oxygen and glucose deprivation treatment. We provide evidence that the neuroprotective activity of PBI-05204 is mediated through oleandrin and/or other cardiac glycoside constituents, but that additional, non-cardiac glycoside components of PBI-05204 may also contribute to the observed neuroprotective activity. Finally, we show directly that both oleandrin and the protective activity of PBI-05204 are blood brain barrier penetrant in a novel model for in vivo neuroprotection. Together, these findings suggest clinical potential for PBI-05204 in the treatment of ischemic stroke and prevention of associated neuronal death.


Assuntos
Cardenolídeos/uso terapêutico , Nerium/química , Fármacos Neuroprotetores/uso terapêutico , Fitoterapia/métodos , Acidente Vascular Cerebral/prevenção & controle , Animais , Glicosídeos Cardíacos/química , Glicosídeos Cardíacos/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/deficiência , Hipóxia/tratamento farmacológico , Técnicas In Vitro , ATPase Trocadora de Sódio-Potássio/metabolismo , Fatores de Tempo , Transfecção/métodos
13.
Neurobiol Dis ; 43(1): 248-56, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21458569

RESUMO

Huntington's disease (HD) is a late-onset, neurodegenerative disease for which there are currently no cures nor disease-modifying treatments. Here we report the identification of several potential anti-inflammatory targets for HD using an ex vivo model of HD that involves the acute transfection of human mutant huntingtin-based constructs into rat brain slices. This model recapitulates key components of the human disease, including the formation of intracellular huntingtin protein (HTT)-containing inclusions and the progressive neurodegeneration of striatal neurons-both occurring within the native tissue context of these neurons. Using this "high-throughput biology" screening platform, we conducted a hypothesis-neutral screen of a collection of drug-like compounds which identified several anti-inflammatory targets that provided neuroprotection against HTT fragment-induced neurodegeneration. The nature of these targets provide further support for non-cell autonomous mechanisms mediating significant aspects of neuropathogenesis induced by mutant HTT fragment proteins.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Doença de Huntington/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Animais , Animais Recém-Nascidos , Anti-Inflamatórios não Esteroides/farmacologia , Corpo Estriado/patologia , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
14.
Hum Mol Genet ; 19(9): 1756-65, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20147317

RESUMO

Huntingtin protein (Htt) is ubiquitously expressed, yet Huntington's disease (HD), a fatal neurologic disorder produced by expansion of an Htt polyglutamine tract, is characterized by neurodegeneration that occurs primarily in the striatum and cerebral cortex. Such discrepancies between sites of expression and pathology occur in multiple neurodegenerative disorders associated with expanded polyglutamine tracts. One possible reason is that disease-modifying factors are tissue-specific. Here, we show that the striatum-enriched protein, CalDAG-GEFI, is severely down-regulated in the striatum of mouse HD models and is down-regulated in HD individuals. In the R6/2 transgenic mouse model of HD, striatal neurons with the largest aggregates of mutant Htt have the lowest levels of CalDAG-GEFI. In a brain-slice explant model of HD, knock-down of CalDAG-GEFI expression rescues striatal neurons from pathology induced by transfection of polyglutamine-expanded Htt exon 1. These findings suggest that the striking down-regulation of CalDAG-GEFI in HD could be a protective mechanism that mitigates Htt-induced degeneration.


Assuntos
Corpo Estriado/metabolismo , Regulação para Baixo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Doença de Huntington/metabolismo , Animais , Corpo Estriado/patologia , Humanos , Proteína Huntingtina , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Ratos , Ratos Sprague-Dawley
15.
J Neurosci ; 28(36): 9013-20, 2008 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-18768695

RESUMO

Although expanded polyglutamine (polyQ) repeats are inherently toxic, causing at least nine neurodegenerative diseases, the protein context determines which neurons are affected. The polyQ expansion that causes Huntington's disease (HD) is in the first exon (HDx-1) of huntingtin (Htt). However, other parts of the protein, including the 17 N-terminal amino acids and two proline (polyP) repeat domains, regulate the toxicity of mutant Htt. The role of the P-rich domain that is flanked by the polyP domains has not been explored. Using highly specific intracellular antibodies (intrabodies), we tested various epitopes for their roles in HDx-1 toxicity, aggregation, localization, and turnover. Three domains in the P-rich region (PRR) of HDx-1 are defined by intrabodies: MW7 binds the two polyP domains, and Happ1 and Happ3, two new intrabodies, bind the unique, P-rich epitope located between the two polyP epitopes. We find that the PRR-binding intrabodies, as well as V(L)12.3, which binds the N-terminal 17 aa, decrease the toxicity and aggregation of HDx-1, but they do so by different mechanisms. The PRR-binding intrabodies have no effect on Htt localization, but they cause a significant increase in the turnover rate of mutant Htt, which V(L)12.3 does not change. In contrast, expression of V(L)12.3 increases nuclear Htt. We propose that the PRR of mutant Htt regulates its stability, and that compromising this pathogenic epitope by intrabody binding represents a novel therapeutic strategy for treating HD. We also note that intrabody binding represents a powerful tool for determining the function of protein epitopes in living cells.


Assuntos
Anticorpos/uso terapêutico , Região Variável de Imunoglobulina/metabolismo , Mutação/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Proteínas Nucleares/metabolismo , Prolina/metabolismo , Animais , Anticorpos/metabolismo , Anticorpos/farmacologia , Especificidade de Anticorpos/fisiologia , Encéfalo/metabolismo , Linhagem Celular Transformada , Modelos Animais de Doenças , Mapeamento de Epitopos , Humanos , Proteína Huntingtina , Região Variável de Imunoglobulina/genética , Técnicas In Vitro , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Penicilamina/análogos & derivados , Penicilamina/metabolismo , Peptídeos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Estrutura Terciária de Proteína , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...