Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 6(10)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34027891

RESUMO

Individuals with heart failure (HF) frequently present with comorbidities, including obesity, insulin resistance, hypertension, and dyslipidemia. Many patients with HF experience cardiogenic dementia, yet the pathophysiology of this disease remains poorly understood. Using a swine model of cardiometabolic HF (Western diet+aortic banding; WD-AB), we tested the hypothesis that WD-AB would promote a multidementia phenotype involving cerebrovascular dysfunction alongside evidence of Alzheimer's disease (AD) pathology. The results provide evidence of cerebrovascular insufficiency coupled with neuroinflammation and amyloidosis in swine with experimental cardiometabolic HF. Although cardiac ejection fraction was normal, indices of arterial compliance and cerebral blood flow were reduced, and cerebrovascular regulation was impaired in the WD-AB group. Cerebrovascular dysfunction occurred concomitantly with increased MAPK signaling and amyloidogenic processing (i.e., increased APP, BACE1, CTF, and Aß40 in the prefrontal cortex and hippocampus) in the WD-AB group. Transcriptomic profiles of the stellate ganglia revealed the WD-AB group displayed an enrichment of gene networks associated with MAPK/ERK signaling, AD, frontotemporal dementia, and a number of behavioral phenotypes implicated in cognitive impairment. These provide potentially novel evidence from a swine model that cerebrovascular and neuronal pathologies likely both contribute to the dementia profile in a setting of cardiometabolic HF.


Assuntos
Amiloide/metabolismo , Transtornos Cerebrovasculares , Insuficiência Cardíaca , Doenças Metabólicas , Animais , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/fisiopatologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/fisiopatologia , Transdução de Sinais , Suínos
2.
Bioorg Chem ; 92: 103194, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31493706

RESUMO

Cathepsin B plays key roles in tumor progression with its overexpression being associated with invasive and metastatic phenotypes and is a primary target of protease activated antibody-directed prodrug therapy. It therefore represents a potential therapeutic and diagnostic target and effort has been made to develop fluorescent probes to report on Cathepsin B activity in cells and animal models of cancer. We have designed, synthesized, and thoroughly evaluated four novel "turn on" probes that employ a lysosomotropic dansylcadaverine dye to report on Cathepsin B activity. Enzyme activity assays using a recombinant human enzyme and cancer cell lysates coupled with confocal microscopy experiments demonstrated that one of the probes, derivatized with the self-immolative prodrug linker p-aminobenzyl alcohol, can selectively report on Cathepsin B in biological samples including live cells.


Assuntos
Cadaverina/análogos & derivados , Catepsina B/análise , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/metabolismo , Neoplasias/diagnóstico por imagem , Compostos de Aminobifenil/química , Cadaverina/síntese química , Cadaverina/metabolismo , Catepsina B/metabolismo , Catepsina L/análise , Catepsina L/metabolismo , Linhagem Celular Tumoral , Humanos , Hidrólise , Cinética , Microscopia Confocal , Estrutura Molecular , Imagem Óptica , Proteínas Recombinantes/análise , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade
3.
Front Cardiovasc Med ; 6: 129, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552273

RESUMO

Post-menopausal women with heart failure (HF) frequently exhibit cardiogenic dementia. Using a pre-clinical swine model of post-menopausal HF, we recently demonstrated that experimental menopause (ovariectomy; OVX) and HF (6-month cardiac pressure overload/aortic banding; AB) independently altered cerebral vasomotor control and together impaired cognitive function. The purpose of this study was to examine the prefrontal cortex and hippocampus tissues from these animals to assess whether OVX and HF are associated with neurologic alterations that may contribute to cardiogenic dementia. We hypothesized that OVX and HF would independently alter neuronal cell signaling in swine with post-menopausal cardiogenic dementia. Immunoblot analyses revealed OVX was associated with reduced estrogen receptor-α in both brain regions and HF tended to exacerbate OVX-induced deficits in the hippocampus. Further, OVX was associated with a reduction in the ratio of phosphorylated:total Akt and ERK in the hippocampus as well as decreased total Akt and synaptophysin in the prefrontal cortex. In contrast, HF was associated with a trend toward reduced phosphorylated:total ERK in the prefrontal cortex. In addition, HF was associated with decreased ß-amyloid (1-38) in the prefrontal cortex and increased ß-amyloid (1-38) in the hippocampus. Regional brain lipid analysis revealed OVX tended to increase total, saturated, and monounsaturated fatty acid content in the prefrontal cortex, with the greatest magnitude of change occurring in the AB-OVX group. The data from this study suggest that OVX and HF are independently associated with regional-specific neurologic changes in the brain that contribute to the cardiogenic dementia profile in this model. This pre-clinical swine model may be a useful tool for better understanding post-menopausal cardiogenic dementia pathology and developing novel therapies.

4.
Mol Oncol ; 13(2): 422-440, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30548174

RESUMO

Chromosomal rearrangements involving the mixed-lineage leukemia (MLL1) gene are common in a unique group of acute leukemias, with more than 100 fusion partners in this malignancy alone. However, do these fusions occur or have a role in solid tumors? We performed extensive network analyses of MLL1-fusion partners in patient datasets, revealing that multiple MLL1-fusion partners exhibited significant interactions with the androgen-receptor signaling pathway. Further exploration of tumor sequence data from TCGA predicts the presence of MLL1 fusions with truncated SET domain in prostate tumors. To investigate the physiological relevance of MLL1 fusions in solid tumors, we engineered a truncated version of MLL1 by fusing it with one of its known fusion partners, ZC3H13, to use as a model system. Functional characterization with cell-based assays revealed that MLL1-ZC3H13 fusion induced chromosomal instability, affected mitotic progression, and enhanced tumorsphere formation. The MLL1-ZC3H13 chimera consistently increased the expression of a cancer stem cell marker (CD44); in addition, we detected potential collateral lethality between DOT1L and MLL1 fusions. Our work reveals that MLL1 fusions are likely prevalent in solid tumors and exhibit a potential pro-tumorigenic role.


Assuntos
Proteínas de Ciclo Celular/genética , Instabilidade Cromossômica/genética , Proteína de Leucina Linfoide-Mieloide/genética , Fusão Oncogênica , Proteínas de Fusão Oncogênica/genética , Proteínas Recombinantes de Fusão/genética , Sequência de Bases , Biomarcadores Tumorais/metabolismo , Carcinogênese/metabolismo , Carcinogênese/patologia , Células Clonais , Redes Reguladoras de Genes , Células HCT116 , Humanos , Modelos Biológicos , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas Nucleares , Proteínas de Fusão Oncogênica/metabolismo , Fenótipo , Proteínas de Ligação a RNA , Proteínas Recombinantes de Fusão/metabolismo
5.
Can J Psychiatry ; 64(1): 18-29, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29685068

RESUMO

The focus on the ß-amyloid (Aß) peptide in clinical Alzheimer disease (AD) as well as in animal models of AD has perhaps biased our understanding of what contributes to the heterogeneity in disease onset and progression. Part of this heterogeneity could reflect the various neuropsychiatric risk factors that present with common symptomatology and can predispose the brain to AD-like changes. One such risk factor is depression. Animal models, particularly mouse models carrying variants of AD-related gene(s), many of which lead to an accumulation of Aß, suggest that a fundamental shift in depression-related monoaminergic systems (including serotonin and noradrenaline) is a strong indicator of the altered cellular function associated with the earlier(est) stages of AD-related pathology. These changes in monoaminergic neurochemistry could provide for relevant targets for intervention in clinical AD and/or could support a polypharmacy strategy, which might include the targeting of Aß, in vulnerable populations. Future studies must also include female mice as well as male mice in animal model studies on the relationship between depression and AD.


Assuntos
Doença de Alzheimer/complicações , Depressão/complicações , Modelos Animais de Doenças , Animais
6.
Oncogene ; 37(30): 4073-4093, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29700392

RESUMO

Triple-negative breast cancer (TNBC) tumours that lack expression of oestrogen, and progesterone receptors, and do not overexpress the HER2 receptor represent the most aggressive breast cancer subtype, which is characterised by the resistance to therapy in frequently relapsing tumours and a high rate of patient mortality. This is likely due to the resistance of slowly proliferating tumour-initiating cells (TICs), and understanding molecular mechanisms that control TICs behaviour is crucial for the development of effective therapeutic approaches. Here, we present our novel findings, indicating that an intrinsically catalytically inactive member of the Eph group of receptor tyrosine kinases, EPHB6, partially suppresses the epithelial-mesenchymal transition in TNBC cells, while also promoting expansion of TICs. Our work reveals that EPHB6 interacts with the GRB2 adapter protein and that its effect on enhancing cell proliferation is mediated by the activation of the RAS-ERK pathway, which allows it to elevate the expression of the TIC-related transcription factor, OCT4. Consistent with this, suppression of either ERK or OCT4 activities blocks EPHB6-induced pro-proliferative responses. In line with its ability to trigger propagation of TICs, EPHB6 accelerates tumour growth, potentiates tumour initiation and increases TIC populations in xenograft models of TNBC. Remarkably, EPHB6 also suppresses tumour drug resistance to DNA-damaging therapy, probably by forcing TICs into a more proliferative, drug-sensitive state. In agreement, patients with higher EPHB6 expression in their tumours have a better chance for recurrence-free survival. These observations describe an entirely new mechanism that governs TNBC and suggest that it may be beneficial to enhance EPHB6 action concurrent with applying a conventional DNA-damaging treatment, as it would decrease drug resistance and improve tumour elimination.


Assuntos
Receptores da Família Eph/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Dano ao DNA/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Nus , Recidiva Local de Neoplasia/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Receptor ErbB-2/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Proteínas ras/metabolismo
7.
Oncotarget ; 7(38): 61544-61561, 2016 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-27557495

RESUMO

Chromosomal Instability (CIN) is regarded as a unifying feature of heterogeneous tumor populations, driving intratumoral heterogeneity. Polo-Like Kinase 1 (PLK1), a serine-threonine kinase that is often overexpressed across multiple tumor types, is one of the key regulators of CIN and is considered as a potential therapeutic target. However, targeting PLK1 has remained a challenge due to the off-target effects caused by the inhibition of other members of the polo-like family. Here we use synthetic dosage lethality (SDL), where the overexpression of PLK1 is lethal only when another, normally non-lethal, mutation or deletion is present. Rather than directly inhibiting PLK1, we found that inhibition of PP2A causes selective lethality to PLK1-overexpressing breast, pancreatic, ovarian, glioblastoma, and prostate cancer cells. As PP2A is widely regarded as a tumor suppressor, we resorted to gene expression datasets from cancer patients to functionally dissect its therapeutic relevance. We identified two major classes of PP2A subunits that negatively correlated with each other. Interestingly, most mitotic regulators, including PLK1, exhibited SDL interactions with only one class of PP2A subunits (PPP2R1A, PPP2R2D, PPP2R3B, PPP2R5B and PPP2R5D). Validation studies and other functional cell-based assays showed that inhibition of PPP2R5D affects both levels of phospho-Rb as well as sister chromatid cohesion in PLK1-overexpressing cells. Finally, analysis of clinical data revealed that patients with high expression of mitotic regulators and low expression of Class I subunits of PP2A improved survival. Overall, these observations point to a context-dependent role of PP2A that warrants further exploration for therapeutic benefits.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Instabilidade Cromossômica/efeitos dos fármacos , Genes Supressores de Tumor/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Proteína Fosfatase 2/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Cantaridina/farmacologia , Cantaridina/uso terapêutico , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Mitose/efeitos dos fármacos , Mutação , Neoplasias/genética , Neoplasias/patologia , Fosforilação , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a Retinoblastoma/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Quinase 1 Polo-Like
8.
Oncotarget ; 7(31): 50027-50042, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27418135

RESUMO

Application of tumor genome sequencing has identified numerous loss-of-function alterations in cancer cells. While these alterations are difficult to target using direct interventions, they may be attacked with the help of the synthetic lethality (SL) approach. In this approach, inhibition of one gene causes lethality only when another gene is also completely or partially inactivated. The EPHB6 receptor tyrosine kinase has been shown to have anti-malignant properties and to be downregulated in multiple cancers, which makes it a very attractive target for SL applications. In our work, we used a genome-wide SL screen combined with expression and interaction network analyses, and identified the SRC kinase as a SL partner of EPHB6 in triple-negative breast cancer (TNBC) cells. Our experiments also reveal that this SL interaction can be targeted by small molecule SRC inhibitors, SU6656 and KX2-391, and can be used to improve elimination of human TNBC tumors in a xenograft model. Our observations are of potential practical importance, since TNBC is an aggressive heterogeneous malignancy with a very high rate of patient mortality due to the lack of targeted therapies, and our work indicates that FDA-approved SRC inhibitors may potentially be used in a personalized manner for treating patients with EPHB6-deficient TNBC. Our findings are also of a general interest, as EPHB6 is downregulated in multiple malignancies and our data serve as a proof of principle that EPHB6 deficiency may be targeted by small molecule inhibitors in the SL approach.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Receptores da Família Eph/metabolismo , Mutações Sintéticas Letais , Neoplasias de Mama Triplo Negativas/metabolismo , Quinases da Família src/metabolismo , Acetamidas/química , Animais , Morte Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Feminino , Corantes Fluorescentes/química , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Imuno-Histoquímica , Indóis/química , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Morfolinas , Piridinas/química , RNA Interferente Pequeno/metabolismo , Sulfonamidas/química , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Leukoc Biol ; 98(3): 313-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25990242

RESUMO

CD8(+) T cells provide protection against pathogens and cancer. After encountering a pathogenic antigen, CD8(+) T cells undergo a triphasic program of rapid proliferation, contraction, and memory formation. Most (∼90-95%) CD8(+) T cells die after vigorous proliferation in the T cell contraction phase, yet the mechanism that triggers apoptotic T cell death remains elusive. This study tested the hypothesis that differential cell-surface expression of M6PR, a multifunctional receptor that regulates lysozyme biogenesis, but also uptakes apoptosis-inducing serine-protease Gzm-B, critically determines life vs. death decisions in T cells. We demonstrate that M6PR-expression on CD8(+) T cell surfaces is dynamically regulated during LmOVA bacterial infection. Notably, time-lapse, confocal microscopy and flow cytometry confirms that M6PR(low) effectors, but not M6PR(high) effectors, escape Gzm-B lethal-hit derived from CD4(+)25(+) Treg cells. Adoptive cotransfer of M6PR(low) effectors and M6PR(high) effectors sorted from LmOVA-infected, congenic mice at the peak of CD8(+) T cell response, reveals that M6PR(low) effectors with the CD8(+) T cell memory precursor phenotype preferentially survive the CD8(+) T cell contraction and differentiate into functional, long-lasting memory CD8(+) T cells. Taken together, our data provide the first evidence, to our knowledge, that selective M6PR down-regulation has a critical role in CD8(+) T cell survival, and our findings have implications for efficient vaccine design and immunotherapy.


Assuntos
Receptor IGF Tipo 2/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Animais , Apoptose , Granzimas/metabolismo , Humanos , Memória Imunológica , Listeria monocytogenes/imunologia , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Perforina/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia
10.
Cell Signal ; 26(12): 2621-32, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25152370

RESUMO

Monoamine oxidase-A (MAO-A) dysfunction has been historically associated with depression. Recently, depression as well as altered MAO-A expression have both been associated with a poor prognosis in cancers, although the mechanism involved remains ambiguous. For example, MAO-A mRNA is repressed across cancers, yet MAO-A protein and levels of serotonin, a substrate of MAO-A implicated in depression, are paradoxically increased in malignancies, including breast cancer. The effect of clorgyline (CLG), a selective inhibitor of MAO-A, on malignant behaviour, expression of transitional markers, and biochemical correlates was examined in two human breast carcinoma cell lines, i.e. the epithelial, oestrogen receptor (ER)-positive MCF-7 cell line and the post-EMT (mesenchymal), ER-negative MDA-MB-231 cell line. CLG exerted little effect on malignant behaviour in MCF-7 cells, but inhibited proliferation and anchorage-independent growth, and increased invasiveness and active migration of MDA-MB-231 cells. CLG induced the expression of the mesenchymal marker vimentin in MCF-7 cells, but not in MDA-MB-231 cells. In contrast, CLG induced the epithelial protein marker E-cadherin in both cell lines, with a more robust effect in MDA-MB-231 cells (where a nuclear E-cadherin signal was also detected). This effect appears to be independent of any canonical Snai1-mediated regulation of E-cadherin mRNA expression. CLG interfered with the ß-catenin/[phospho]GSK-3ß complex as well as the E-cadherin/ß-catenin complex in both cell lines cells, but, again, the effect was more robust in MDA-MB-231 cells. Parallel studies revealed a general lack of effect of CLG on the ER-negative, epithelial Au565 breast cancer cell line. Thus, any effect of CLG on metastatic behaviours appears to rely on the cell's EMT status rather than on the cell's ER status. These data suggest that inactivation of MAO-A triggers a mesenchymal-to-epithelial transition in MDA-MB-231 cells via a non-canonical mechanism. This potentially implicates an MAO-A-sensitive step in advanced breast cancer and should be borne in mind when considering pharmacological treatment options for co-morbid depression in breast cancer patients.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Clorgilina/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Monoaminoxidase/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Células MCF-7 , Invasividade Neoplásica/genética , RNA Mensageiro/genética , Vimentina/metabolismo , beta Catenina/metabolismo
11.
J Neural Transm (Vienna) ; 119(11): 1285-94, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22382901

RESUMO

Post-translational influences could underlie the ambiguous roles of monoamine oxidase-A (MAO-A) in pathologies such as depression, cancer and Alzheimer disease. In support of this, we recently demonstrated that the Ca²âº-sensitive component of MAO-A catalytic activity is inhibited by a pro-survival p38 (MAPK)-dependent mechanism. We substituted three aspartic acid (D) residues in human MAO-A that reside in putative Ca²âº-binding motifs and overexpressed the individual proteins in the human HEK293 cell line. We assayed the overexpressed proteins for catalytic activity and for their influence on cell viability (using MTT conversion and trypan blue exclusion) and proliferation/DNA synthesis [using bromodeoxyuridine (BrdU) incorporation]. Innate MAO-A catalytic activity (and the capacity for generating hydrogen peroxide) was unaffected by the D61A substitution, but inhibited moderately or completely by the D248A and D328G substitutions, respectively. The Ca²âº-sensitive activities of wild-type and D248A MAO-A proteins were enhanced by treatment with the selective p38(MAPK) inhibitor, SB203580, but was completely abrogated by the D61A substitution. Monoamine oxidase-A(D61A) was toxic to cells and exerted no effect on cell proliferation, while MAO-A(D248A) was generally comparable to wild-type MAO-A. As expected, the catalytic-dead MAO-A(D328G) was not cytotoxic, but unexpectedly enhanced both MTT conversion and BrdU staining. Variant-dependent changes in Bax and Bcl-2/Bcl-XL protein expression were observed. A different pattern of effects in N2-a cells suggests cell line-dependent roles for MAO-A. A catalytic-dependent mechanism influences MAO-A-mediated cytotoxicity, whereas a catalytic-independent mechanism contributes to proliferation. Context-dependent inputs by either mechanism could underlie the ambiguous pathological contributions of MAO-A.


Assuntos
Ácido Aspártico/metabolismo , Proliferação de Células/efeitos dos fármacos , Monoaminoxidase/metabolismo , Mutação/genética , Análise de Variância , Animais , Bromodesoxiuridina/metabolismo , Cálcio/farmacologia , Catálise/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Inibidores Enzimáticos/farmacologia , Humanos , Imidazóis/farmacologia , Imunoprecipitação , Camundongos , Monoaminoxidase/genética , Mutagênese Sítio-Dirigida/métodos , Neuroblastoma/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Piridinas/farmacologia , Serotonina/farmacocinética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Transfecção , Trítio/farmacocinética , Proteína bcl-X/metabolismo
12.
J Alzheimers Dis ; 28(2): 403-22, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22045496

RESUMO

The concentration of presenilin-1 (PS-1) protein at the mitochondrial-associated aspect of the endoplasmic reticulum supports the potential for a mitochondrial influence of PS-1. Given that carriers of certain Alzheimer's disease (AD)-related PS-1 variants are predisposed to clinical depression and that depression has been historically associated with the mitochondrial enzyme, monoamine oxidase-A (MAO-A), we investigated cortical MAO-A function in the AD-related PS-1(M146V) knock-in mouse. The MAO-A system was clearly altered in the PS-1(M146V) mouse as revealed by (a) a mismatch between MAO-A protein expression and MAO-A activity; (b) changes in MAO-A-mediated monoaminergic neurotransmitter metabolism; (c) changes in non-cognitive behavior following treatment with the irreversible MAO-A inhibitor clorgyline; and (d) an increase in the potency of clorgyline in these same mice. We next investigated whether PS-1(M146V) could be influencing MAO-A directly. We observed (a) an enhanced MAO-A activity in necropsied PS-1(M146V) mouse cortical extracts incubated with DAPT (a PS-1 substrate-competitor); (b) the proximity of PS-1 with MAO-A and mitochondrial markers in cortical sections and in primary cortical neurons; (c) the co-segregation and co-immunoprecipitation of PS-1 and MAO-A within the mitochondrial fraction; and (d) the co-immunoprecipitation of overexpressed PS-1(M146V) and MAO-A proteins from N2a lysates. The PS-1(ΔEx9) and PS-1(D257A) variants, known to have low substrate-binding capacity, co-immunoprecipitated weakly with MAO-A. These combined data support a physical interaction between PS-1 and MAO-A that could influence MAO-A activity and contribute to the monoaminergic disruptions common to disorders as seemingly diverse as depression and AD.


Assuntos
Doença de Alzheimer/patologia , Córtex Cerebral/enzimologia , Regulação Enzimológica da Expressão Gênica/genética , Metionina/genética , Monoaminoxidase/metabolismo , Presenilina-1/genética , Valina/genética , Doença de Alzheimer/genética , Análise de Variância , Animais , Células Cultivadas , Córtex Cerebral/patologia , Cromatografia Líquida de Alta Pressão/métodos , Clorgilina/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Ácido Hidroxi-Indolacético/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Inibidores da Monoaminoxidase/farmacologia , Mutação/genética , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/ultraestrutura , Neurotransmissores/metabolismo , Compostos Orgânicos , Serotonina/metabolismo , Frações Subcelulares/enzimologia , Natação/psicologia , Transfecção , Canais de Ânion Dependentes de Voltagem/metabolismo
13.
J Neural Transm (Vienna) ; 118(7): 987-95, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21373759

RESUMO

Monoamine oxidase-A (MAO-A) has been associated with both depression and Alzheimer disease (AD). Recently, carriers of AD-related presenilin-1 (PS-1) alleles have been found to be at higher risk for developing clinical depression. We chose to examine whether PS-1 could influence MAO-A function in vitro. Overexpression of selected AD-related PS-1 variants (wildtype, Y115H, ΔEx9 and M146V) in mouse hippocampal HT-22 cells affects MAO-A catalytic activity in a variant-specific manner. The ability of the PS-1 substrate-competitor DAPT to induce MAO-A activity in cells expressing either PS-1 wildtype or PS-1(M146V) suggests the potential for a direct influence of PS-1 on MAO-A function. In support of this, we were able to co-immunoprecipitate MAO-A with FLAG-tagged PS-1 wildtype and M146V proteins. This potential for a direct protein-protein interaction between PS-1 and MAO-A is not specific for HT-22 cells as we were also able to co-immunoprecipitate MAO-A with FLAG-PS-1 variants in N2a mouse neuroblastoma cells and in HEK293 human embryonic kidney cells. Finally, we demonstrate that the two PS-1 variants reported to be associated with an increased incidence of clinical depression [e.g., A431E and L235V] both induce MAO-A activity in HT-22 cells. A direct influence of PS-1 variants on MAO-A function could provide an explanation for the changes in monoaminergic tone observed in several neurodegenerative processes including AD. The ability to induce MAO-A catalytic activity with a PS-1/γ-secretase inhibitor should also be considered when designing secretase inhibitor-based therapeutics.


Assuntos
Doença de Alzheimer/enzimologia , Transtorno Depressivo/enzimologia , Variação Genética , Monoaminoxidase/metabolismo , Neurônios/enzimologia , Presenilina-1/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Transtorno Depressivo/genética , Transtorno Depressivo/patologia , Células HEK293 , Humanos , Camundongos , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Neurônios/citologia , Presenilina-1/fisiologia
14.
FASEB J ; 25(5): 1746-57, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21321187

RESUMO

Methylglyoxal (MG), a reactive dicarbonyl molecule, can modify protein to form advanced glycation endproducts. Increased MG level has been implicated in proliferative vascular diseases, but the underlying mechanisms are not clear yet. The serine/threonine kinase, Akt, regulates multiple signaling pathways that control cell proliferation. Using mass spectrometric analysis, we have detected the modification of Akt1 by MG at Cys(77). This structural modification increased Akt1 phosphorylation at Ser(473) and Thr(308). Akt1 phosphorylation and activity were also increased by MG treatment (<50 µM) in cultured vascular smooth muscle cells (VSMCs). MG treatment of VSMCs led to increased DNA synthesis (EC(50)=5.8 µM), cell proliferation, phosphorylation of p21 and glycogen synthase kinase-3α/ß (GSK-3α/ß), and increased cyclin-dependent kinase 2 (CDK2) activity. These effects of MG were significantly inhibited by silencing Akt1 or by an Akt inhibitor. Overexpression of Akt1 Cys(77)Ser mutant in HEK-293 cells increased cell proliferation and DNA synthesis, concurrent with an increase in Akt1 activity, which could not be further augmented by MG treatment. It is concluded that MG-induced VSMC proliferation is mediated by the activation of Akt1 via the modification of Akt1 at Cys(77).


Assuntos
Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Aldeído Pirúvico/farmacologia , Animais , Western Blotting , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células HEK293 , Humanos , Masculino , Espectrometria de Massas , Miócitos de Músculo Liso/citologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Ratos
15.
Life Sci ; 85(5-6): 262-8, 2009 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-19539632

RESUMO

AIMS: Calcium (Ca(2+)) is known to augment monoamine oxidase-A (MAO-A) activity in cell cultures as well as in brain extracts from several species. This association between Ca(2+) and MAO-A could contribute to their respective roles in cytotoxicity. However, the effect of Ca(2+) on MAO-A function in human brain has as yet to be examined as does the contribution of specific signalling cascades. MAIN METHODS: We examined the effects of Ca(2+) on MAO-A activity and on [(3)H]Ro 41-1049 binding to MAO-A in human cerebellar extracts, and compared this to its effects on MAO-A activity in glial C6 cells following the targeting of signalling pathways using specific chemical inhibitors. KEY FINDINGS: Ca(2+) enhances MAO-A activity as well as the association of [(3)H]Ro 41-1049 to MAO-A in human cerebellar extracts. The screening of neuronal and glial cell cultures reveals that MAO-A activity does not always correlate with the expression of either mao-A mRNA or MAO-A protein. Inhibition of the individual PI3K/Akt, ERK and p38(MAPK) signalling pathways in glial C6 cells all augment basal MAO-A activity. Inhibition of the p38(MAPK) pathway also augments Ca(2+)-sensitive MAO-A activity. We also observe the inverse relation between p38(MAPK) activation and MAO-A function in C6 cultures grown to full confluence. SIGNIFICANCE: The Ca(2+)-sensitive component to MAO-A activity is present in human brain and in vitro studies link it to the p38(MAPK) pathway. This means of influencing MAO-A function could explain its role in pathologies as diverse as neurodegeneration and cancers.


Assuntos
Cálcio/farmacologia , Cerebelo/efeitos dos fármacos , Monoaminoxidase/metabolismo , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Idoso , Animais , Sítios de Ligação , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Cerebelo/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Glioblastoma , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Monoaminoxidase/genética , Inibidores da Monoaminoxidase/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Ligação Proteica , RNA Mensageiro/metabolismo , Ratos , Tiazóis/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Cell Signal ; 21(1): 161-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18951975

RESUMO

The antipsychotic drug haloperidol is still used to treat psychosis and "agitation", often with devastating consequences, particularly in geriatric and pre-demented patients. Cytotoxicity induced by haloperidol has been associated with induction of Bcl-XS, a pro-apoptotic member of the Bcl-2 family, as well as with modulation of the Akt pro-survival pathway. Using preneuronal PC12 and primary neuronal cultures, we show that haloperidol inactivates Akt. This induces the dephosphorylation of serine residues in Bcl-XS and promotes its association with the mitochondrial voltage-dependent anion channel (VDAC), as well as with cytochrome c- and caspase-3-dependent events. These events are sensitive to expression of constitutively active Akt. Mutation of Serine106 (Ser106), which is flanked by a putative Akt motif, hinders the association of the Bcl-XS protein with Akt, but promotes its association with VDAC. The dephosphorylation mimic, Bcl-XS(Ser106Ala), induces caspase-dependent PC12 and neuronal cell apoptosis. In contrast, Bcl-XS(Ser106Ala) induces a significant loss of VDAC expression, and cytochrome c- and caspase-independent toxicity in the non-neuronal HEK293A cells. We link haloperidol and Akt to Bcl-XS-sensitive toxicity via cell line-dependent mitochondrial events centering on VDAC. This clearly mitigates the chronic use of haloperidol in neuropsychiatric populations, but supports its use as a potential acute therapeutic in cancer, where apoptosis is desirable.


Assuntos
Antipsicóticos/toxicidade , Haloperidol/toxicidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína bcl-X/metabolismo , Análise de Variância , Animais , Apoptose , Caspase 3/metabolismo , Linhagem Celular , Citocromos c/metabolismo , Haloperidol/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mutagênese Sítio-Dirigida , Células PC12 , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Transdução de Sinais , Canais de Ânion Dependentes de Voltagem/metabolismo , Canais de Ânion Dependentes de Voltagem/fisiologia
17.
Cell Signal ; 20(11): 2038-49, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18718528

RESUMO

Protein kinase B (PKB; also known as Akt) is important for mediating survival and proliferation signals. Following activation, PKB shuttles to various compartments of the cell, including the nucleus, where it phosphorylates an array of targets. PKB is phosphorylated at T308 by its activator PDK1. PDK1 is normally excluded from the nucleus via a nuclear exclusion sequence (NES), and our previous work suggested that nuclear exclusion can be attenuated by IGF-1-induced phosphorylation of S396 proximal to the NES. No studies have been done to test the significance of S396 phosphorylation or the impact of nuclear accumulation of PDK1 on PKB activation. To address these questions, we created isogenic embryonic stem cell (ESC) lines expressing various alleles of PDK1 within a PDK1-/- background. Disruption of the NES domain of PDK1 correlated with elevated PKB phosphorylation at both T308 and S473. In contrast, mutation of S396 to alanine reduced PDK1 nuclear localization and reduced PKB phosphorylation and activation. The loss of phosphorylation of PKB by S396A mutation was rescued by forcing nuclear PDK1 or by conversion of S396 to an aspartic acid. The phosphorylation of the PKB substrate FOXO3alpha was reduced in S396A PDK1 ESC. Other known and suspected PKB substrates, including GSK3 and Raf1, were unaffected. This study therefore reveals that S396 plays a role in the activation of PKB leading to the regulated phosphorylation of some PKB substrates including FOXO3alpha.


Assuntos
Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Motivos de Aminoácidos , Animais , Linhagem Celular , Células Clonais , Células-Tronco Embrionárias/enzimologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Mutação/genética , Fosforilação , Estrutura Terciária de Proteína , Transporte Proteico , Recombinases/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
18.
J Psychiatry Neurosci ; 32(5): 323-30, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17823648

RESUMO

OBJECTIVE: The antipsychotic drug haloperidol (HAL) has been linked to apoptosis and to inhibition of prosurvival Akt signalling in pheochromocytoma (PC12) and neuronal cell cultures. However, the mechanism involved is unclear. METHODS: We used HAL to induce cytotoxicity in preneuronal PC12 cells. The expression and the subcellular localization of selected components of the PI3K-Akt survival cascade were monitored with standard biochemical approaches, such as subcellular fractionation, western blot analysis, gene transfer and fluorescence microscopy. RESULTS: PC12 cell stimulation with the epidermal growth factor (used as a control) results in normal processing of phosphatidylinositol 3'-kinase (PI3K)-Akt signalling (e.g., localization of PI3K to the plasma membrane and phosphorylation of Akt (Ser473). Surprisingly, HAL induces PI3K-generated phosphoinositol [phosphatidylinositol-3,4,5-triphosphate (PIP3), which conflicts with its ability to inhibit Akt. In fact, the production of PIP3s is nuclear, as assessed by the localized concentration of a fluorophore-tagged PIP3-targeting pleckstrin homology protein and a fluorophore-tagged substrate-trapping mutant of the phosphoinositide phosphatase, phosphatase and tensin homologue deleted on chromosome 10 (PTEN). However, phosphoinositide-dependent protein kinase 1 (PDK1, the activating kinase of Akt) does not colocalize to the nucleus with the PI3K complex. This effectively inactivates both cytoplasmic and nuclear pools of Akt. CONCLUSION: The differential compartmentalization of effectors of the PI3K-PDK1-Akt pathway is a unique means by which HAL disrupts Akt functioning in PC12 cells.


Assuntos
Haloperidol/farmacologia , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Translocação Genética/efeitos dos fármacos , Animais , Clonagem Molecular , DNA Complementar/biossíntese , DNA Complementar/genética , Células PC12 , Fosforilação , Ratos , Transfecção
19.
J Neurosci Res ; 74(6): 942-7, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14648600

RESUMO

We have demonstrated recently that atypical antipsychotics possess neuroprotective actions in H2O2-mediated and serum-withdrawal models of cell death. In the present study, we compared the ability of atypical and typical antipsychotics to protect against an insult mediated by Abeta(25-35), an apoptogenic fragment of the Alzheimer's disease-related beta-amyloid (Abeta) peptide. Treatment of PC12 cell cultures with Abeta(25-35) did not significantly alter total cellular expression levels of Bax, a proapoptotic Bcl-2 family member, or levels of Bcl-XL, an antiapoptotic analogue. Treatment with Abeta(25-35), however, did result in mitochondrial translocation of Bax, which effectively increased the mitochondrial ratio of Bax to Bcl-X(L). This relative increase in proapoptotic molecules was reduced by pretreatment with atypical (quetiapine and olanzapine) and typical (haloperidol) antipsychotics. We also observed a selective increase in proapoptotic Bcl-XS immunodetection in haloperidol-treated cells, which was evident particularly in the mitochondrial compartment. This increase in proapoptotic molecules may account for the lower neuroprotective potential of haloperidol, as determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT) reduction assay. The disparate neuroprotective effects of atypical and typical antipsychotics/neuroleptics may be due to their respective abilities to regulate pro- and anti-apoptotic protein translocation and expression.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Antipsicóticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Regulação da Expressão Gênica/fisiologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células PC12 , Fragmentos de Peptídeos/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Ratos , Proteína X Associada a bcl-2 , Proteína bcl-X
20.
J Biol Chem ; 278(40): 38715-22, 2003 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-12867422

RESUMO

Transforming growth factor-beta (TGF-beta) receptor-mediated signaling has been proposed to mediate both the beneficial and deleterious roles for this cytokine in amyloid-beta protein (Abeta) function. In order to assess receptor dependence of these events, we used PC12 cell cultures, which are devoid of TGF-beta receptors. Surprisingly, TGF-beta potentiated the neurotoxic effects of the 40-residue Abeta peptide, Abeta-(1-40), in this model suggesting that there may be a direct, receptor-independent interaction between TGF-beta and Abeta-(1-40). Surface plasmon resonance confirmed that TGF-beta binds with high affinity directly to Abeta-(1-40) and electron microscopy revealed that TGF-beta enhances Abeta-(1-40) oligomerization. Immunohistochemical examination of mouse brain revealed that hippocampal CA1 and dentate gyrus, two regions classically associated with Abeta-mediated pathology, lack TGF-beta Type I receptor immunoreactivity, thus indicating that TGF-beta receptor-mediated signaling would not be favored in these regions. Our observations not only provide for a unique, receptor-independent mechanism of action for TGF-beta, but also help to reconcile the literature interpreting the role of TGF-beta in Abeta function. These data support a critical etiological role for this mechanism in neuropathological amyloidoses.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Ligação Competitiva , Linhagem Celular , Dicroísmo Circular , Densitometria , Feminino , Hipocampo/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Neurônios/metabolismo , Células PC12 , Ligação Proteica , Isoformas de Proteínas , Ratos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA