Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Neuroepidemiology ; 2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38295773

RESUMO

Native American individuals are more frequently affected by cerebrovascular diseases including stroke and vascular cognitive decline. The aim of this study is to determine stroke risk factors that are most prevalent in Wisconsin Native Americans and to examine how education at the community and individual level as well as intensive health wellness coaching may influence modification of stroke risk factors. Additionally, we will investigate the role novel stroke biomarkers may play in stroke risk in this population. This paper details the aims and methods employed in the "Stroke Prevention in the Wisconsin Native American Population" (clinicaltrials.gov identifier: NCT04382963) study including participant health assessments, clinical ultrasound exam of the carotid arteries, cognitive testing battery and structure and execution of the coaching program.

2.
Neurochem Int ; 164: 105499, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36746322

RESUMO

Cerebrovascular disease (CVD) is the second most common cause of cognitive impairment and dementia in aged population. CVD presents in a myriad number of clinical ways based on the functional location of pathology. While primary clinical emphasis has been placed on motor, speech and visual deficits, vascular cognitive decline is a vastly under recognized and devastating condition afflicting millions of Americans. CVD, a disease of the blood vessels that supply blood to brain involves an integration between small and large vessels. Cerebral large vessel diseases (LVD) are associated with atherosclerosis, artery-to-artery embolism, intracardiac embolism and a large vessel stroke leading to substantial functional disability. Cerebral small vessel disease (SVD) is critically involved in stroke, brain hemorrhages, cognitive decline and functional loss in elderly patients. An evolving understanding of cellular and molecular mechanisms emphasizes that inflammatory vascular changes contribute to systemic pathologic conditions of the central nervous systems (CNS), with specific clinical presentations including, cognitive decline. Advances in an understanding of pathophysiology of disease processes and therapeutic interventions may help improve outcomes. This review will focus on large and small vessels diseases and their relationship to vascular cognitive decline, atherosclerosis, stroke, and inflammatory neurodegeneration. We will also emphasize the molecular and cellular mechanisms, as well as genetic and epigenetic factors associated with LVD and SVD.


Assuntos
Aterosclerose , Doenças de Pequenos Vasos Cerebrais , Transtornos Cerebrovasculares , Acidente Vascular Cerebral , Idoso , Humanos , Transtornos Cerebrovasculares/patologia , Acidente Vascular Cerebral/patologia , Encéfalo/patologia , Doenças de Pequenos Vasos Cerebrais/patologia , Aterosclerose/complicações , Aterosclerose/patologia
3.
Brain Res ; 1776: 147752, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34906547

RESUMO

Glioblastoma (GB) is highly invasive and resistant to multimodal treatment partly due to distorted vasculature and exacerbated inflammation. The aggressiveness of brain tumors may be attributed to the dysregulated release of angiogenic and inflammatory factors. The glycoprotein pentraxin-3 (PTX3) is correlated with the severity of some cancers. However, the mechanism responsible for the invasive oncogenic role of PTX3 in GB malignancy remains unclear. In this study, we examined the role of PTX3 in GB growth, angiogenesis, and invasion using in vitro and in vivo GB models, proteomic profiling, molecular and biochemical approaches. Under in vitro conditions, PTX3 over-expression in U87 cells correlated with cell cycle progression, increased migratory potential, and proliferation under hypoxic conditions. Conditioned media containing PTX3 enhanced the angiogenic potential of endothelial cells. While silencing of PTX3 by siRNA decreased the proliferation, migration, and angiogenic potential of U87 cells in vitro. Importantly, PTX3 over-expression increased tumor growth, angiogenesis, and invasion in an orthotopic mouse model. Higher levels of PTX3 in these tumors were associated with the upregulation of inflammatory and angiogenic markers including interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF), but decreased levels of thrombospondin-1, an anti-angiogenic factor. Mechanistically, exogenous production of PTX3 triggered an IKK/NFκB signaling pathway that enhances the expression of the motility genes AHGEF7 and Rac1. Taken together, PTX3 expression is dysregulated in GB. PTX3 may augment invasion through enhanced angiogenesis in the GB microenvironment through the IL8-VEGF axis. Thus, PTX3 may represent a potential therapeutic target to mitigate the aggressive behavior of gliomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Proteína C-Reativa/metabolismo , Glioblastoma/metabolismo , Interleucina-8/metabolismo , Invasividade Neoplásica/genética , Componente Amiloide P Sérico/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proteína C-Reativa/genética , Linhagem Celular , Glioblastoma/genética , Glioblastoma/patologia , Camundongos , Invasividade Neoplásica/patologia , Neurônios/metabolismo , Neurônios/patologia , Componente Amiloide P Sérico/genética
4.
J Cereb Blood Flow Metab ; 41(4): 857-873, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33736511

RESUMO

Post-stroke neurological deficits and mortality are often associated with vascular disruption and neuronal apoptosis. Galectin-3 (Gal3) is a potent pro-survival and angiogenic factor. However, little is known about its protective role in the cerebral ischemia/reperfusion (I/R) injury. We have previously shown significant up-regulation of Gal3 in the post-stroke rat brain, and that blocking of Gal3 with neutralizing antibody decreases the cerebral blood vessel density. Our current study demonstrates that intracerebral local delivery of the Gal3 into rat brain at the time of reperfusion exerts neuroprotection. Ischemic lesion volume and neuronal cell death were significantly reduced as compared with the vehicle-treated MCAO rat brains. Gal3 increased vessel density and neuronal survival after I/R in rat brains. Importantly, Gal3-treated groups showed significant improvement in motor and sensory functional recovery. Gal3 increased neuronal cell viability under in vitro oxygen-glucose deprivation conditions in association with increased phosphorylated-Akt, decreased phosphorylated-ERK1/2, and reduced caspase-3 activity. Gene expression analysis showed down regulation of pro-apoptotic and inflammatory genes including Fas-ligand, and upregulation of pro-survival and pro-angiogenic genes including Bcl-2, PECAM, and occludin. These results indicate a key role for Gal3 in neuro-vascular protection and functional recovery following ischemic stroke through modulation of angiogenic and apoptotic pathways.


Assuntos
Indutores da Angiogênese/farmacologia , Apoptose/efeitos dos fármacos , Caspases/efeitos dos fármacos , Galectina 3/uso terapêutico , AVC Isquêmico/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Encéfalo , Morte Celular/efeitos dos fármacos , Galectina 3/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Hipóxia Encefálica/tratamento farmacológico , Microinjeções , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos , Ratos Endogâmicos SHR , Traumatismo por Reperfusão/prevenção & controle
5.
Neurochem Int ; 127: 113-124, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30707914

RESUMO

Metabolic dysfunction impacts stroke incidence and outcome. However, the intricate association between altered metabolic program due to aging, and focal ischemia in brain, circulation, and peripheral organs is not completely elucidated. Here we identified locally and systemically altered metabolites in brain, liver, and plasma as a result of normal aging, ischemic-stroke, and extended time of reperfusion injury. Comprehensive quantitative metabolic profiling was carried out using nuclear magnetic resonance spectroscopy. Aging, but healthy rats showed significant metabolic alterations in the brain, but only a few metabolic changes in the liver and plasma as compared to younger rats. But, ischemic stroke altered metabolites significantly in liver and plasma of older rats during early acute phase. Major metabolic changes were also seen in the brains of younger rats following ischemic stroke during early acute phase of injury. We further report that metabolic changes occur sequentially in a tissue specific manner during extended reperfusion time of late repair phase. First metabolic alterations occurred in brain due to local injury. Next, changes in circulating metabolites in plasma occurred during acute-repair phase transition time. Lastly, the delayed systemic effect was seen in the peripheral organ, liver that exhibited significant and persistent changes in selected metabolites during later reperfusion time. The metabolic pathways involved in energy/glucose, and amino acid metabolism, inflammation, and oxidative stress were mainly altered as a result of aging and ischemia/reperfusion. Biomarker analysis revealed citrate, lysine, and tyrosine as potential age-independent blood metabolic biomarkers of ischemia/reperfusion. Overall, our study elucidates the complex network of metabolic events as a function of normal aging and acute stroke. We further provide evidence for a clear transition from local to systemic metabolic dysfunction due to ischemic injury in a time dependent manner, which may altogether greatly impact the post-stroke outcome.


Assuntos
Envelhecimento/fisiologia , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Fígado/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Espectroscopia de Ressonância Magnética/métodos , Metabolômica/métodos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Traumatismo por Reperfusão/metabolismo
6.
Mol Neurobiol ; 54(7): 4973-4985, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27525674

RESUMO

Cerebral ischemia evokes abnormal release of proteases in the brain microenvironment that spatiotemporally impact angio-neurogenesis. Dipeptidyl peptidase IV (DPPIV), a cell surface and secreted protease, has been implicated in extracellular matrix remodeling by regulating cell adhesion, migration, and angiogenesis through modifying the functions of the major chemokine stromal-derived factor, SDF1. To elucidate the possible association of DPPIV in ischemic brain, we examined the expression of DPPIV in the post-stroke rat brain and under in vitro ischemia by oxygen glucose deprivation (OGD). We further investigated the effects of DPPIV on SDF1 mediated in vitro chemotactic and angiogenic functions. DPPIV protein and mRNA levels were significantly upregulated during repair phase in the ischemic cortex of the rat brain, specifically in neurons, astrocytes, and endothelial cells. In vitro exposure of Neuro-2a neuronal cells and rat brain endothelial cells to OGD resulted in upregulation of DPPIV. In vitro functional analysis showed that DPPIV decreases the SDF1-mediated angiogenic potential of rat brain endothelial cells and inhibits the migration of Neuro-2a and neural progenitor cells. Western blot analyses revealed decreased levels of phosphorylated ERK1/2 and AKT in the presence of DPPIV. DPPIV inhibitor restored the effects of SDF1. Proteome profile array screening further revealed that DPPIV decreases matrix metalloproteinase-9, a key downstream effector of ERK-AKT signaling pathways. Overall, delayed induction of DPPIV in response to ischemia/reperfusion suggests that DPPIV may play an important role in endogenous brain tissue remodeling and repair processes. This may be mediated through modulation of SDF1-mediated cell migration and angiogenesis.


Assuntos
Isquemia Encefálica/enzimologia , Movimento Celular/fisiologia , Dipeptidil Peptidase 4/metabolismo , Neovascularização Patológica/metabolismo , Células-Tronco Neurais/enzimologia , Acidente Vascular Cerebral/enzimologia , Animais , Encéfalo/metabolismo , Isquemia Encefálica/patologia , Movimento Celular/efeitos dos fármacos , Células Endoteliais/metabolismo , Masculino , Neurogênese/fisiologia , Ratos , Acidente Vascular Cerebral/terapia
7.
Ann Neurosci ; 23(1): 25-32, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27536019

RESUMO

Neuropathic pain (NP) affects approximately 4 million people in the United States with spinal cord injury (SCI) being a common cause. Matrix metalloproteinases (MMPs) play an integral role in mediating inflammatory responses, cellular signaling, cell migration, extracellular matrix degradation and tissue remodeling and repair. As such, they are major components in the pathogenesis of secondary injury within the central nervous system. Other gene regulatory pathways, specifically MAPK/extracellular signaling-regulated kinase (ERK) and Wnt/ß-catenin, are also believed to participate in secondary injury likely intersect. The study aims to examine the MMP-2 signaling pathway associated with ERK and Wnt/ß-catenin activity during contusion SCI (cSCI)-induced NP in a rat model. This is an experimental study investigating the implication of MMP-2 in SCI-induced NP and its association with the cellular and molecular changes in the interactions between extracellular signaling kinase and ß-catenin. Adult Sprague-Dawley rats received cSCI injury by NYU impactor by dropping 10 g weight from a height of 12.5 mm. Locomotor functional recovery of injured rats was measured on post cSCI day 1, and weekly thereafter for 6 weeks using Basso, Beattie and Bresnahan scores. Thermal hyperalgesia (TH) testing was performed on days 21, 28, 35 and 42 post cSCI. The expression and/or activity of MMP-2, ß-catenin and ERK were studied following harvest of spinal cord tissues between 3 and 6 weeks post cSCI. All experiments were funded by the department of Neurological Surgery at the University of Wisconsin, School of Medicine and Public Health having no conflict of interest. MMP-2 and ß-catenin expression were elevated and gradually increased from days 21 to 42 compared to sham-operated rats and injured rats that did not exhibit TH. The expression of phosphorylated ERK (phospho-ERK) increased on day 21 but returned to baseline levels on day 42 whereas total ERK levels remained relatively unchanged and constant. Chronic NP is associated with changes in the expression of MMP-2, ß-catenin and ERK. Our data suggest that the transient upregulation of phospho-ERK is involved in the initial upregulation of both ß-catenin and MMP-2 following cSCI-induced NP states.

8.
Mol Neurobiol ; 51(3): 1530-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25084761

RESUMO

Sphingomyelin synthase (SMS) is a key enzyme involved in the generation of sphingomyelin (SM) and regulation of cell growth and survival. However, the effects of SMS on neuronal cell proliferation and cell cycle progression are not completely elucidated. In this study, we examined the direct effects of SMS1 in regulating cell cycle progression and proliferation of Neuro-2a cells that exhibit neuronal characteristics. Neuro-2a cells transfected with SMS-specific small hairpin RNA (shRNA) expressed significantly lower levels of SMS1. RNA interference-mediated depletion of SMS1 in Neuro-2a cells caused a significant decrease in SM levels. Decreased SMS1 levels resulted in reduced proliferation rate and morphological changes including neurite-like outgrowth. Also, silencing of SMS1 induced cell cycle arrest as shown by the increased percentage of cells in G0/G1 and decreased proportion of cells in S phase. These changes were accompanied by upregulation of cyclin-dependent kinase inhibitor p27 and decreased levels of cyclin D1 and phospho-Akt. Nuclear accumulation of p27 was also evident in SMS1-deficient cells. Furthermore, loss of SMS1 inhibited the migratory potential of Neuro-2a cells in association with decreased levels of matrix metalloproteinases. These results indicate that SMS1 plays an important role in mediating the key signaling pathways that are involved in the tight coordination of multiple cellular activities, including neuronal cell proliferation, cell cycle progression, and migration, and therefore may have significant implications in neurodegenerative diseases.


Assuntos
Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Proliferação de Células/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo , Animais , Linhagem Celular , Camundongos , Transdução de Sinais/fisiologia
9.
PLoS One ; 8(1): e54391, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23349873

RESUMO

The respiratory epithelium is subject to continuous environmental stress and its responses to injury or infection are largely mediated by transactivation of the epidermal growth factor receptor (EGFR) and downstream signaling cascades. Based on previous studies indicating involvement of ATP-dependent activation of the NADPH oxidase homolog DUOX1 in epithelial wound responses, the present studies were performed to elucidate the mechanisms by which DUOX1-derived H(2)O(2) participates in ATP-dependent redox signaling and EGFR transactivation. ATP-mediated EGFR transactivation in airway epithelial cells was found to involve purinergic P2Y(2) receptor stimulation, and both ligand-dependent mechanisms as well as ligand-independent EGFR activation by the non-receptor tyrosine kinase Src. Activation of Src was also essential for ATP-dependent activation of the sheddase ADAM17, which is responsible for liberation and activation of EGFR ligands. Activation of P2Y(2)R results in recruitment of Src and DUOX1 into a signaling complex, and transient siRNA silencing or stable shRNA transfection established a critical role for DUOX1 in ATP-dependent activation of Src, ADAM17, EGFR, and downstream wound responses. Using thiol-specific biotin labeling strategies, we determined that ATP-dependent EGFR transactivation was associated with DUOX1-dependent oxidation of cysteine residues within Src as well as ADAM17. In aggregate, our findings demonstrate that DUOX1 plays a central role in overall epithelial defense responses to infection or injury, by mediating oxidative activation of Src and ADAM17 in response to ATP-dependent P2Y(2)R activation as a proximal step in EGFR transactivation and downstream signaling.


Assuntos
Proteínas ADAM/metabolismo , Receptores ErbB/genética , Infecções , NADPH Oxidases/metabolismo , Quinases da Família src/metabolismo , Proteínas ADAM/genética , Proteína ADAM17 , Trifosfato de Adenosina/metabolismo , Células Cultivadas , Oxidases Duais , Receptores ErbB/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Infecções/genética , Infecções/metabolismo , NADPH Oxidases/genética , Oxirredução , Fosforilação , Receptores Purinérgicos P2Y/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Transdução de Sinais , Ativação Transcricional , Cicatrização/genética , Cicatrização/fisiologia , Quinases da Família src/genética
10.
Brain Res ; 1496: 1-9, 2013 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-23246924

RESUMO

Focal cerebral ischemia initiates self-repair mechanisms that include the production of neurotrophic factors and cytokines. Galectin-3 is an important angiogenic cytokine. We have previously demonstrated that expression of galectin 3 (Gal-3), a carbohydrate binding protein is significantly upregulated in activated microglia in the brains of rats subjected to focal ischemia. Further blocking of Gal-3 function with Gal-3 neutralizing antibody decreased the microvessel density in ischemic brain. We currently show that Gal-3 significantly increases the viability of microglia BV2 cells subjected to oxygen glucose deprivation (OGD) and re-oxygenation. Exogenous Gal-3 promoted the formation of pro-angiogenic structures in an in vitro human umbilical vein endothelial (HUVEC) and BV2 cell co-culture model. Gal-3 induced angiogenesis was associated with increased expression of vascular endothelial growth factor. The conditioned medium of BV2 cells exposed to OGD contained increased Gal-3 levels, and promoted the formation of pro-angiogenic structures in an in vitro HUVEC culture model. Gal-3 also augmented the in vitro migratory potential of BV2 microglia. Gal-3 mediated functions were associated with increased levels of integrin-linked kinase (ILK) signaling as demonstrated by the impaired angiogenesis and migration of BV2 cells following targeted silencing of ILK expression by siRNA. Furthermore, we show that ILK levels correlate with the levels of phos-AKT and ERK1/2 that are downstream effectors of ILK pathway. Taken together, our studies indicate that Gal-3 contributes to angiogenesis and microglia migration that may have implications in post stroke repair.


Assuntos
Movimento Celular/efeitos dos fármacos , Galectina 3/farmacologia , Integrinas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Microglia/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Contagem de Células , Linhagem Celular Transformada , Movimento Celular/fisiologia , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Galectina 3/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glucose/deficiência , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Camundongos , Microglia/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/farmacologia , Fatores de Tempo
11.
Mol Neurobiol ; 45(3): 455-64, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22415444

RESUMO

Tricyclodecan-9-yl-xanthogenate (D609) inhibits phosphatidylcholine (PC)-phospholipase C (PLC) and/or sphingomyelin (SM) synthase (SMS). Inhibiting SMS can increase ceramide levels, which can inhibit cell proliferation. Here, we examined how individual inflammatory and glia cell proliferation is altered by D609. Treatment with 100-µM D609 significantly attenuated the proliferation of RAW 264.7 macrophages, N9 and BV-2 microglia, and DITNC(1) astrocytes, without affecting cell viability. D609 significantly inhibited BrdU incorporation in BV-2 microglia and caused accumulation of cells in G(1) phase with decreased number of cells in the S phase. D609 treatment for 2 h significantly increased ceramide levels in BV-2 microglia, which, following a media change, returned to control levels 22 h later. This suggests that the effect of D609 may be mediated, at least in part, through ceramide increase via SMS inhibition. Western blots demonstrated that 2-h treatment of BV-2 microglia with D609 increased expression of the cyclin-dependent kinase (Cdk) inhibitor p21 and down-regulated phospho-retinoblastoma (Rb), both of which returned to basal levels 22 h after removal of D609. Exogenous C8-ceramide also inhibited BV-2 microglia proliferation without loss of viability and decreased BrdU incorporation, supporting the involvement of ceramide in D609-mediated cell cycle arrest. Our current data suggest that D609 may offer benefit after stroke (Adibhatla and Hatcher, Mol Neurobiol 41:206-217, 2010) through ceramide-mediated cell cycle arrest, thus restricting glial cell proliferation.


Assuntos
Hidrocarbonetos Aromáticos com Pontes/farmacologia , Ciclo Celular/efeitos dos fármacos , Ceramidas/metabolismo , Tionas/farmacologia , Animais , Bromodesoxiuridina/metabolismo , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ceramidas/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Diglicerídeos/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Camundongos , Norbornanos , Esfingomielinas/metabolismo , Tiocarbamatos , Fatores de Tempo
12.
PLoS One ; 6(7): e21876, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21750738

RESUMO

Polypyrimidine Tract Binding (PTB) protein is a regulator of mRNA processing and translation. Genetic screens and studies of wing and bristle development during the post-embryonic stages of Drosophila suggest that it is a negative regulator of the Notch pathway. How PTB regulates the Notch pathway is unknown. Our studies of Drosophila embryogenesis indicate that (1) the Notch mRNA is a potential target of PTB, (2) PTB and Notch functions in the dorso-lateral regions of the Drosophila embryo are linked to actin regulation but not their functions in the ventral region, and (3) the actin-related Notch activity in the dorso-lateral regions might require a Notch activity at or near the cell surface that is different from the nuclear Notch activity involved in cell fate specification in the ventral region. These data raise the possibility that the Drosophila embryo is divided into zones of different PTB and Notch activities based on whether or not they are linked to actin regulation. They also provide clues to the almost forgotten role of Notch in cell adhesion and reveal a role for the Notch pathway in cell fusions.


Assuntos
Actinas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Receptores Notch/genética , Actinas/metabolismo , Animais , Apoptose/genética , Northern Blotting , Adesão Celular/genética , Fusão Celular , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Mutação , Miocárdio/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/genética
13.
Int J Cancer ; 129(6): 1322-30, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21128244

RESUMO

Neuroblastoma, a cancer of the sympathetic nervous system, is the most common extracranial solid tumor in children. MYCN amplification and increased BDNF/TrkB signaling are features of high-risk tumors; yet, only ˜25% of malignant tumors display these features. Thus, the identification of additional biomarkers and therapeutic targets is essential. As aminoacylase 1 (ACY1), an amino acid deacetylase, is a putative tumor suppressor in small cell lung and renal cell carcinomas, we investigated whether it or the other family members aspartoacylase (ASPA, aminoacylase 2) or aminoacylase 3 (ACY3) could serve a similar function in neuroblastoma. Aminoacylase expression was examined in TrkB-positive, MYCN-amplified (SMS-KCNR and SK-N-BE) and TrkB-negative, non-MYCN-amplified (SK-N-AS, SK-N-SH, SH-SY5Y and SH-EP) neuroblastoma cell lines. Each aminoacylase exhibited distinct spatial localization (i.e., cytosolic ACY1, membrane-associated ASPA and nuclear ACY3). When SK-N-SH cells were treated with neural differentiation agents (e.g., retinoic acid and cAMP) in media containing 10% serum, ACY1 was the only aminoacylase whose expression was upregulated. ASPA was primarily expressed in SH-EP cells of a glial sublineage. ACY3 was more highly expressed in the TrkB-positive, MYCN-amplified lines. All three aminoacylases were expressed in normal human adrenal gland, a common site of neuroblastoma origin, but only ACY1 and ACY3 displayed detectable expression in primary neuroblastoma tumor. Bioinformatics data mining of Kaplan-Meier survival revealed that high ACY3 expression is correlated with poor prognosis, whereas low expression of ACY1 or ASPA is correlated with poor prognosis. These data suggest that aminoacylase expression is dysregulated in neuroblastoma.


Assuntos
Neoplasias das Glândulas Suprarrenais/enzimologia , Glândulas Suprarrenais/enzimologia , Amidoidrolases/metabolismo , Neuroblastoma/enzimologia , Adolescente , Adulto , Diferenciação Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neuroblastoma/patologia
14.
J Biol Chem ; 283(26): 17919-28, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18424783

RESUMO

Increased synthesis of NO during airway inflammation, caused by induction of nitric-oxide synthase 2 in several lung cell types, may contribute to epithelial injury and permeability. To investigate the consequence of elevated NO production on epithelial function, we exposed cultured monolayers of human bronchial epithelial cells to the NO donor diethylenetriaamine NONOate. At concentrations generating high nanomolar levels of NO, representative of inflammatory conditions, diethylenetriaamine NONOate markedly reduced wound closure in an in vitro scratch injury model, primarily by inhibiting epithelial cell migration. Analysis of signaling pathways and gene expression profiles indicated a rapid induction of the mitogen-activated protein kinase phosphatase (MPK)-1 and decrease in extracellular signal-regulated kinase (ERK)1/2 activation, as well as marked stabilization of hypoxia-inducible factor (HIF)-1alpha and activation of hypoxia-responsive genes, under these conditions. Inhibition of ERK1/2 signaling using U0126 enhanced HIF-1alpha stabilization, implicating ERK1/2 dephosphorylation as a contributing mechanism in NO-mediated HIF-1alpha activation. Activation of HIF-1alpha by the hypoxia mimic cobalt chloride, or cell transfection with a degradation-resistant HIF-1alpha mutant construct inhibited epithelial wound repair, implicating HIF-1alpha in NO-mediated inhibition of cell migration. Conversely, NO-mediated inhibition of epithelial wound closure was largely prevented after small interfering RNA suppression of HIF-1alpha. Finally, NO-mediated inhibition of cell migration was associated with HIF-1alpha-dependent induction of PAI-1 and activation of p53, both negative regulators of epithelial cell migration. Collectively, our results demonstrate that inflammatory levels of NO inhibit epithelial cell migration, because of suppression of ERK1/2 signaling, and activation of HIF-1alpha and p53, with potential consequences for epithelial repair and remodeling during airway inflammation.


Assuntos
Células Epiteliais/citologia , Regulação Enzimológica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Inflamação , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Movimento Celular , Ativação Enzimática , Humanos , Modelos Biológicos , RNA Interferente Pequeno/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
15.
Front Biosci ; 13: 2435-43, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981724

RESUMO

Dipeptidyl peptidase IV (DPPIV), a serine protease is expressed by normal melanocytes but not by melanomas, the malignant counterpart. DPPIV is encoded by a gene that contains a 5 CpG island spanning a transcriptional regulatory region. Previously we have demonstrated that DPPIV abrogates growth factor independence and functions as a tumor suppressor gene in melanomas. In this study we show that loss of DPPIV occurs at RNA level and demethylating agent, 5-aza-2'-deoxycytidine (5-AZA-Cdr) treatment of DPPIV negative melanoma cell lines results in increase of DPPIV mRNA, protein, and enzyme activities. By using sodium bisulfite genomic DNA modifications, PCR, and sequencing we confirmed that DPPIV gene promoter is methylated in eight out of ten melanoma cell lines tested. Further more, 5-AZA-Cdr induced increases in DPPIV levels correlated with growth inhibition and apoptosis in melanoma cells. All together these findings suggest that frequent downregulation of DPPIV expression in melanoma can be attributed, in large part, to aberrant promoter hypermethylation and this loss of DPPIV may be a critical event contributing to melanoma development.


Assuntos
Metilação de DNA , Dipeptidil Peptidase 4/fisiologia , Inativação Gênica , Genes Supressores de Tumor , Melanoma/metabolismo , Regiões Promotoras Genéticas , Neoplasias Cutâneas/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Dipeptidil Peptidase 4/metabolismo , Progressão da Doença , Humanos , Melanoma/genética , Neoplasias Cutâneas/genética
16.
Am J Respir Cell Mol Biol ; 36(2): 138-46, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16980554

RESUMO

The airway epithelium provides a protective barrier against inhaled environmental toxins and microorganisms, and epithelial injury initiates a number of processes to restore its barrier integrity, including activation of matrix metalloproteinases such as MMP-9 (92-kD gelatinase B). Airway epithelial cells continuously produce nitric oxide (NO), which has been linked to cell migration and MMP-9 regulation in several cell types, but the importance of epithelial NO in mediating airway epithelial repair or MMP-9 activation is unknown. Using primary or immortalized human bronchial epithelial cells, we demonstrate that low concentrations of NO promote epithelial cell migration and wound repair in an in vitro wound assay, which was associated with increased localized expression and activation of MMP-9. In addition, in HBE1 cells that were stably transfected with inducible NOS (NOS2), to mimic constitutive epithelial NOS2 expression in vivo, NOS inhibition decreased epithelial wound repair and MMP-9 expression. The stimulatory effects of NO on epithelial wound repair and MMP-9 expression were dependent on cGMP-mediated pathways and were inhibited by 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ), an inhibitor of soluble guanylyl cyclase. Inhibition of cGMP-dependent protein kinase (PKG) attenuated NO-mediated epithelial wound closure, but did not affect MMP-9 expression. However, pharmacologic MMP inhibition and siRNA knockdown of MMP-9 expression demonstrated the contribution of MMP-9 to NO-mediated wound closure. Overall, our results demonstrate that NOS2-derived NO contributes to airway epithelial repair by both PKG-dependent and -independent mechanisms, and involves NO-dependent expression and activation of MMP-9.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Óxido Nítrico/farmacologia , Sistema Respiratório/citologia , Sistema Respiratório/enzimologia , Cicatrização/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Gelatinases/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Metaloproteinase 9 da Matriz/genética , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Compostos Nitrosos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , ômega-N-Metilarginina/farmacologia
17.
J Biol Chem ; 282(5): 3213-20, 2007 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-17135261

RESUMO

The airway epithelium is continuously subjected to environmental pollutants, airborne pathogens, and allergens and relies on several intrinsic mechanisms to maintain barrier integrity and to promote epithelial repair processes following injury. Here, we report a critical role for dual oxidase 1 (Duox1), a newly identified NADPH oxidase homolog within the tracheobronchial epithelium, in airway epithelial cell migration and repair following injury. Activation of Duox1 during epithelial injury is mediated by cellular release of ATP, which signals through purinergic receptors expressed on the epithelial cell surface. Purinergic receptor stimulation by extracellular ATP is a critical determinant of epithelial cell migration and repair following injury and is associated with activation of extracellular signal-regulated kinases (ERK1/2) and matrix metalloproteinase-9 (MMP-9). Stimulation of these integral features of epithelial cell migration and repair processes was found to require the activation of Duox1. Our findings demonstrate a novel role for Duox1 in the tracheobronchial epithelium, in addition to its proposed role in antimicrobial host defense, by participating in epithelial repair processes to maintain epithelial integrity and barrier function in the face of environmental stress.


Assuntos
Trifosfato de Adenosina/metabolismo , Flavoproteínas/metabolismo , NADPH Oxidases/metabolismo , Mucosa Respiratória/fisiologia , Cicatrização , Brônquios , Primers do DNA , Oxidases Duais , Humanos , Peróxido de Hidrogênio/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Traqueia
18.
Dev Dyn ; 235(2): 411-26, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16331645

RESUMO

Notch signaling is used to specify cell types during animal development. A high level specifies one cell type, whereas a low level specifies the alternate type. The effector of Notch signaling is the Notch intracellular domain. Upon its release from the plasma membrane in response to Delta binding the Notch extracellular domain, the Notch intracellular domain combines with the transcription factor Suppressor of Hairless and promotes the expression of target genes. Using a panel of antibodies made against different extracellular and intracellular regions of Notch, we show that cell types and tissues with low levels of Notch signaling are enriched for Notch molecules detected only by the extracellular domain antibodies. This enrichment often follows enrichment for Notch molecules detected only by antibodies made against the Suppressor of Hairless binding region. Notch molecules lacking most of the intracellular domain or containing only the Suppressor of Hairless binding region are produced during development. Such molecules are known to suppress Notch signaling, possibly by taking away Delta or Suppressor of Hairless from the full-length Notch. Thus, it is possible that dominant-negative Notch molecules are produced in the normal course of tissue differentiation in Drosophila as part of an auto-down-regulation mechanism.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Genes Dominantes/genética , Mutação/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Animais , Animais Geneticamente Modificados , Anticorpos/imunologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Proteínas de Drosophila/imunologia , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Epitopos/imunologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Receptores Notch/imunologia , Transdução de Sinais/genética
19.
Cancer Res ; 65(4): 1325-34, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15735018

RESUMO

Dipeptidyl peptidase IV (DPPIV) is a serine protease with tumor suppressor function. It regulates the activities of mitogenic peptides implied in cancer development. Progression of benign prostate cancer to malignant metastasis is linked to increased production of basic fibroblast growth factor (bFGF), a powerful mitogen. In this study, using in vitro model system we show that DPPIV loss is associated with increased bFGF production in metastatic prostate cancer cells. DPPIV reexpression in prostate cancer cells blocks nuclear localization of bFGF, reduces bFGF levels, inhibits mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK)1/2 activation, and decreases levels of urokinase-type plasminogen activator, known downstream effectors of bFGF signaling pathway. These molecular changes were accompanied by induction of apoptosis, cell cycle arrest, inhibition of in vitro cell migration, and invasion. Silencing of DPPIV by small interfering RNA resulted in increased bFGF levels and restoration of mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK)1/2 activation. These results indicate that DPPIV inhibits the malignant phenotype of prostate cancer cells by blocking bFGF signaling pathway.


Assuntos
Adenosina Desaminase/fisiologia , Dipeptidil Peptidase 4/fisiologia , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Glicoproteínas/fisiologia , Neoplasias da Próstata/enzimologia , Adenosina Desaminase/biossíntese , Adenosina Desaminase/deficiência , Adenosina Desaminase/genética , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Dipeptidil Peptidase 4/biossíntese , Dipeptidil Peptidase 4/genética , Ativação Enzimática , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibronectinas/biossíntese , Glicoproteínas/biossíntese , Glicoproteínas/deficiência , Glicoproteínas/genética , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Fosforilação , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
20.
Int J Cancer ; 109(6): 855-66, 2004 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15027119

RESUMO

Lung cancer is the leading cause of cancer death. Lung cancers produce a variety of mitogenic growth factors that stimulate tumor cell proliferation and migration. The cell surface protease, dipeptidyl peptidase IV (DPPIV), is involved in diverse biologic functions, including peptide-mediated cellular growth and differentiation. DPPIV is expressed in various normal tissues, including lung tissue, and its expression is lost in many types of human cancers. DPPIV expression and its enzymatic activity are detected in normal bronchial and alveolar epithelium but different histologic subtypes of lung carcinomas lose DPPIV expression. To investigate the role of DPPIV in lung carcinoma, we examined the expression of DPPIV at both mRNA and protein levels in non small cell lung cancer (NSCLC) cell lines and normal human bronchial epithelial cells. DPPIV expression was detectable in normal lung epithelial cells, but was absent or markedly reduced in all NSCLC cell lines at both mRNA and protein levels. Restoration of DPPIV expression in NSCLC cells resulted in profound morphologic changes, inhibition of cell proliferation, anchorage-independent growth, in vitro cell migration and tumorigenicity in nude mice. DPPIV reexpression also correlated with increased p21 expression, leading to induction of apoptosis and cell cycle arrest in G1 stage. These effects were accompanied by increased expression of cell surface proteins, fibroblast-activating protein (Fapalpha) and CD44 that are associated with suppression of tumor growth and metastasis. Thus, DPPIV functions as a tumor suppressor, and its downregulation may contribute to the loss of growth control in NSCLC cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/enzimologia , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Gelatinases , Regulação Enzimológica da Expressão Gênica/fisiologia , Neoplasias Pulmonares/enzimologia , Animais , Apoptose , Carcinoma Pulmonar de Células não Pequenas/patologia , Adesão Celular , Movimento Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Regulação para Baixo , Endopeptidases , Perfilação da Expressão Gênica , Humanos , Receptores de Hialuronatos/metabolismo , Marcação In Situ das Extremidades Cortadas , Neoplasias Pulmonares/patologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Serina Endopeptidases/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA