Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Alzheimers Dis ; 35(4): 777-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23542865

RESUMO

Alzheimer's disease (AD) is a global health crisis with limited treatment options. Despite major advances in neurotherapeutics, poor brain penetration due to the blood-brain barrier continues to pose a big challenge in overcoming the access of therapeutics to the central nervous system. In that regard, the non-invasive intranasal route of brain targeting is gaining considerable attention. The nasal mucosa offers a large surface area, rapid absorption, and avoidance of first-pass metabolism increasing drug bioavailability with less systemic side effects. Intranasal delivery is known to utilize olfactory, rostral migratory stream, and trigeminal routes to reach the brain. This investigation confirmed that intranasal delivery of oligomeric amyloid-ß antibody (NU4) utilized all three routes to enter the brain with a resident time of 96 hours post single bolus intranasal administration, and showed evidence of perikaryal and parenchymal uptake of NU4 in 5XFAD mouse brain, confirming the intranasal route as a non-invasive and efficient way of delivering therapeutics to the brain. In addition, this study demonstrated that intranasal delivery of NU4 antibody lowered cerebral amyloid-ß and improved spatial learning in 5XFAD mice.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/imunologia , Anticorpos/metabolismo , Anticorpos/uso terapêutico , Encéfalo/metabolismo , Administração Intranasal , Animais , Anticorpos/administração & dosagem , Cognição/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Nervo Trigêmeo/efeitos dos fármacos
2.
Gene ; 466(1-2): 1-15, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20627123

RESUMO

Dynamic modification involving small ubiquitin-like modifier (SUMO) has emerged as a new mechanism of protein regulation in mammalian biology. Sumoylation is an ATP-dependent, reversible post-translational modification which occurs under both basal and stressful cellular conditions. Sumoylation profoundly influences protein functions and pertinent biological processes. For example, sumoylation modulates multiple components in the NFkappaB pathway and exerts an anti-inflammatory effect. Likewise, sumoylation of peroxisome proliferator-activated receptor gamma (PPARgamma) augments its anti-inflammatory activity. Current evidence suggests a role of sumoylation for resistance to apoptosis in synovial fibroblasts. Dynamic SUMO regulation controls the biological outcomes initiated by various growth factors involved in cartilage homeostasis, including basic fibroblast growth factors (bFGF or FGF-2), transforming growth factor-beta (TGF-beta) and insulin-like growth factor-1 (IGF-1). The impact of these growth factors on cartilage are through sumoylation-dependent control of the transcription factors (e.g., Smad, Elk-1, HIF-1) that are key regulators of matrix components (e.g., aggrecan, collagen) or cartilage-degrading enzymes (e.g., MMPs, aggrecanases). Thus, SUMO modification appears to profoundly affect chondrocyte and synovial fibroblast biology, including cell survival, inflammatory responses, matrix metabolism and hypoxic responses. More recently, evidence suggests that, in addition to their nuclear roles, the SUMO pathways play crucial roles in mitochondrial activity, cellular senescence, and autophagy. With an increasing number of reports linking SUMO to human diseases like arthritis, it is probable that novel and equally important functions of the sumoylation pathway will be elucidated in the near future.


Assuntos
Artrite/genética , Animais , Autofagia , Senescência Celular , Condrócitos/metabolismo , Humanos , Inflamação/genética , Modelos Biológicos , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Membrana Sinovial/metabolismo
3.
Arthritis Rheum ; 62(10): 2995-3005, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20556813

RESUMO

OBJECTIVE: To verify the biologic links between progressive cellular and structural alterations within knee joint components and development of symptomatic chronic pain that are characteristic of osteoarthritis (OA), and to investigate the molecular basis of alterations in nociceptive pathways caused by OA-induced pain. METHODS: An animal model of knee joint OA pain was generated by intraarticular injection of mono-iodoacetate (MIA) in Sprague-Dawley rats, and symptomatic pain behavior tests were performed. Relationships between development of OA with accompanying pain responses and gradual alterations in cellular and structural knee joint components (i.e., cartilage, synovium, meniscus, subchondral bone) were examined by histologic and immunohistologic analysis, microscopic examination, and microfocal computed tomography. Progressive changes in the dynamic interrelationships between peripheral knee joint tissue and central components of nociceptive pathways caused by OA-induced pain were examined by investigating cytokine production and expression in sensory neurons of the dorsal root ganglion and spinal cord. RESULTS: We observed that structural changes in components of the peripheral knee joint correlate with alterations in the central compartments (dorsal root ganglia and the spinal cord) and symptomatic pain assessed by behavioral hyperalgesia. Our comparative gene expression studies revealed that the pain pathways in MIA-induced knee OA may overlap, at least in part, with neuropathic pain mechanisms. Similar results were also observed upon destabilization of the knee joint in the anterior cruciate ligament transection and destabilization of the medial meniscus models of OA. CONCLUSION: Our results indicate that MIA-induced joint degeneration in rats generates an animal model that is suitable for mechanistic and pharmacologic studies on nociceptive pain pathways caused by OA, and provide key in vivo evidence that OA pain is caused by central sensitization through communication between peripheral OA nociceptors and the central sensory system. Furthermore, our data suggest a mechanistic overlap between OA-induced pain and neuropathic pain.


Assuntos
Artralgia/fisiopatologia , Gânglios Espinais/fisiopatologia , Articulação do Joelho/inervação , Nociceptores/fisiologia , Osteoartrite do Joelho/fisiopatologia , Medula Espinal/fisiopatologia , Animais , Artrite Experimental , Condrócitos/metabolismo , Condrócitos/patologia , Osteoartrite do Joelho/patologia , Ratos , Ratos Sprague-Dawley
4.
Neoplasia ; 9(5): 427-34, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17534448

RESUMO

Following genotoxic stress, transcriptional activation of target genes by p53 tumor suppressor is critical in cell fate determination. Here we report that the restoration of p53 function in human cancer cell lines that are deficient in p53 function upregulated the expression of Notch1. Interestingly, the expression of wild-type p53 in human prostate and breast cancer cell lines correlated well with increased expression of Notch1. Furthermore, knockdown of p53 expression in cancer cells that express wild-type p53 resulted in reduced expression of Notch1. Importantly, genotoxic stress to cancer cells that resulted in activation of p53 also upregulated the expression of Notch1. Moreover, p53-mediated induction of Notch1 expression was associated with stimulation of the activity of Notch-responsive reporters. Notably, p53 differentially regulated the expression of Notch family members: expression of Notch2 and Notch4 was not induced by p53. Significantly, treatment of cells with gamma secretase inhibitor, an inhibitor of Notch signaling, increased susceptibility to apoptosis in response to genotoxic stress. Together, our observations suggest that p53-mediated upregulation of Notch1 expression in human cancer cell lines contributes to cell fate determination after genotoxic stress.


Assuntos
Dano ao DNA , Regulação Neoplásica da Expressão Gênica , Neoplasias/patologia , Receptor Notch1/genética , Proteína Supressora de Tumor p53/fisiologia , Apoptose , Linhagem Celular Tumoral , Etoposídeo/farmacologia , Feminino , Humanos , Masculino , Neoplasias/genética , RNA Mensageiro/análise , Regulação para Cima
5.
Mol Cancer Res ; 5(3): 251-9, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17339605

RESUMO

Increased expression of IFI16 protein (encoded by the IFI16 gene) in normal human prostate epithelial cells is associated with cellular senescence-associated cell growth arrest. Consistent with a role for IFI16 protein in cellular senescence, the expression of IFI16 protein is either very low or not detectable in human prostate cancer cell lines. We now report that treatment of DU-145 and LNCaP prostate cancer cell lines with histone deacetylase inhibitor trichostatin A (TSA) or CGK1026 resulted in transcriptional activation of the IFI16 gene. The induction of IFI16 protein in LNCaP cells was dependent on the duration of TSA treatment. Furthermore, TSA treatment of LNCaP cells up-regulated the expression of Janus-activated kinase 1 protein kinase and modulated the transcription of certain IFN-activatable genes. However, overexpression of exogenous Janus-activated kinase 1 protein in LNCaP cells and treatment of cells with IFNs (alpha and gamma) did not increase the expression of IFI16. Instead, the transcriptional activation of IFI16 gene by TSA treatment of LNCaP cells was dependent on transcriptional activation by c-Jun/activator protein-1 transcription factor. Importantly, increased expression of IFI16 in LNCaP cells was associated with decreases in the expression of androgen receptor and apoptosis of cells. Conversely, knockdown of IFI16 expression in TSA-treated LNCaP cells increased androgen receptor protein levels with concomitant decreases in apoptosis. Together, our observations provide support for the idea that histone deacetylase-dependent transcriptional silencing of the IFI16 gene in prostate epithelial cells contributes to the development of prostate cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Histona Desacetilases/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas/genética , Neoplasias da Próstata/genética , Linhagem Celular Tumoral , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Inativação Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Humanos , Ácidos Hidroxâmicos/farmacologia , Janus Quinase 1/metabolismo , Masculino , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Neoplasias da Próstata/enzimologia , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ativação Transcricional , Regulação para Cima
6.
Am J Physiol Heart Circ Physiol ; 288(3): H1477-90, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15388503

RESUMO

Histone deacetylases (HDACs) are a family of enzymes that catalyze the removal of acetyl groups from core histones, resulting in change of chromatin structure and gene transcription activity. In the heart, HDACs are targets of hypertrophic signaling, and their nonspecific inhibition by trichostatin A (TSA) attenuates hypertrophy of cultured cardiac myocytes. In this study, we examined the effect of TSA on two major determinants of cardiac contractility: alpha-myosin heavy chain (MHC) expression and microtubular composition and organization. TSA upregulated the expression of alpha-MHC in cultured cardiac myocytes, as well as in an in vivo model of hypothyroid rats. Studies designed to delineate mechanisms of alpha-MHC induction by TSA revealed an obligatory role of early growth response factor-1 on activation of the alpha-MHC promoter. Concurrently, TSA downregulated the expression of alpha- and beta-tubulins and prevented the induction of tubulins by a hypertrophy agonist, ANG II. The ANG II-mediated increased proportion of alpha- and beta-tubulins associated with polymerized microtubules was also markedly reduced after treatment of cells by TSA. Results obtained from immunofluorescent microscopy indicated that TSA had no noticeable effect on the organization of cardiac microtubules in control cells, whereas it prevented the ANG II-induced dense parallel linear arrays of microtubules to a profile similar to that of controls. Together, these results demonstrate that inhibition of HDACs by TSA regulates the cardiac alpha-MHC and tubulins in a manner predictive of improved cardiac contractile function. These studies improve our understanding of the role of HDACs on cardiac hypertrophy with implications in development of new therapeutic agents for treatment of cardiac abnormalities.


Assuntos
Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Contração Miocárdica/efeitos dos fármacos , Cadeias Pesadas de Miosina/genética , Tubulina (Proteína)/genética , Acetilação/efeitos dos fármacos , Animais , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce , Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Proteínas Imediatamente Precoces/metabolismo , Masculino , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Ratos , Ratos Sprague-Dawley , Fator de Resposta Sérica/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos
7.
J Biol Chem ; 278(22): 20047-58, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12663674

RESUMO

Serum response factor (SRF) plays a pivotal role in cardiac myocyte development, muscle gene transcription, and hypertrophy. Previously, elevation of intracellular levels of Ca2+ was shown to activate SRF function without involving the Ets family of tertiary complex factors through an unknown regulatory mechanism. Here, we tested the hypothesis that the chromatin remodeling enzymes of class II histone deacetylases (HDAC4) regulate SRF activity in a Ca2+-sensitive manner. Expression of HDAC4 profoundly repressed SRF-mediated transcription in both muscle and nonmuscle cells. Protein interaction studies demonstrated physical association of HDAC4 with SRF in living cells. The SRF/HDAC4 co-association was disrupted by treatment of cells with hypertrophic agonists such as angiotensin-II and a Ca2+ ionophore, ionomycin. Furthermore, activation of Ca2+/calmodulin-dependent protein kinase (CaMK)-IV prevented SRF/HDAC4 interaction and derepressed SRF-dependent transcription activity. The SRF.HDAC4 complex was localized to the cell nucleus, and the activated CaMK-IV disrupted HDAC4/SRF association, leading to export of HDAC4 from the nucleus and stimulation of SRF transcription activity. Thus, these results identify SRF as a functional interacting target of HDAC4 and define a novel tertiary complex factor-independent mechanism for SRF activation by Ca2+/CaMK-mediated signaling.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cardiomegalia/metabolismo , Histona Desacetilases/metabolismo , Miocárdio/metabolismo , Proteínas Repressoras/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Sequência de Bases , Cardiomegalia/enzimologia , Células Cultivadas , Sondas de DNA , Microscopia Confocal , Miocárdio/enzimologia , Ratos
8.
Am J Physiol Heart Circ Physiol ; 282(4): H1521-33, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11893590

RESUMO

Serum response factor (SRF) has been shown to play a key role in cardiac cell growth and muscle gene regulation. To understand the role of SRF in heart failure, we compared its expression pattern between control and failing human heart samples. Western blot analysis of control samples showed expression of four different isoforms of SRF, with ~67-kDa full-length SRF being the predominant isoform. Interestingly, in failing hearts we found robust expression of a low-molecular-mass (~52 kDa) SRF isoform, accompanied by decreased expression of full-length SRF. By RT-PCR and Southern blot analyses, we characterized this ~52-kDa SRF isoform as being encoded by an alternatively spliced form of SRF lacking exons 4 and 5 of the SRF primary RNA transcript (SRF-Delta4,5 isoform). We cloned SRF-Delta4,5 cDNA and showed that overexpression of this isoform into cells inhibits SRF-dependent activation of cardiac muscle genes. These results suggest that expression of SRF-Delta4,5 in failing hearts may in part contribute to impaired cardiac gene expression and consequently to the pathogenesis of heart failure.


Assuntos
Processamento Alternativo , Insuficiência Cardíaca/genética , Fator de Resposta Sérica/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Animais , Sequência de Bases , Divisão Celular , Células Cultivadas , Criança , Clonagem Molecular , Primers do DNA , Sondas de DNA , Modelos Animais de Doenças , Éxons , Feminino , Regulação da Expressão Gênica , Insuficiência Cardíaca/patologia , Implante de Prótese de Valva Cardíaca , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Miocárdio/citologia , Miocárdio/metabolismo , Isoformas de Proteínas/genética , Coelhos , Ratos , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Ativação Transcricional , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...