Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Commun Biol ; 4(1): 753, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34140629

RESUMO

Metastatic cancer cells are frequently deficient in WWOX protein or express dysfunctional WWOX (designated WWOXd). Here, we determined that functional WWOX-expressing (WWOXf) cells migrate collectively and expel the individually migrating WWOXd cells. For return, WWOXd cells induces apoptosis of WWOXf cells from a remote distance. Survival of WWOXd from the cell-to-cell encounter is due to activation of the survival IκBα/ERK/WWOX signaling. Mechanistically, cell surface epitope WWOX286-299 (repl) in WWOXf repels the invading WWOXd to undergo retrograde migration. However, when epitope WWOX7-21 (gre) is exposed, WWOXf greets WWOXd to migrate forward for merge. WWOX binds membrane type II TGFß receptor (TßRII), and TßRII IgG-pretreated WWOXf greet WWOXd to migrate forward and merge with each other. In contrast, TßRII IgG-pretreated WWOXd loses recognition by WWOXf, and WWOXf mediates apoptosis of WWOXd. The observatons suggest that normal cells can be activated to attack metastatic cancer cells. WWOXd cells are less efficient in generating Ca2+ influx and undergo non-apoptotic explosion in response to UV irradiation in room temperature. WWOXf cells exhibit bubbling cell death and Ca2+ influx effectively caused by UV or apoptotic stress. Together, membrane WWOX/TßRII complex is needed for cell-to-cell recognition, maintaining the efficacy of Ca2+ influx, and control of cell invasiveness.


Assuntos
Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/patologia , Neoplasias/patologia , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Oxidorredutase com Domínios WW/metabolismo , Animais , Apoptose/imunologia , Células COS , Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Chlorocebus aethiops , Células HCT116 , Humanos , Células L , Células MCF-7 , Camundongos , Inibidor de NF-kappaB alfa/metabolismo , Neoplasias/genética , Transdução de Sinais/fisiologia , Oxidorredutase com Domínios WW/genética
2.
Neurosci Lett ; 451(2): 139-43, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19135129

RESUMO

Acoustic stimulation is known to induce c-Fos expression in the auditory system but how the expression might be related to the time-variance of the sound (e.g., steady or frequency-varying) is unclear. Here we measured morphometrically Fos-immunohistochemical stains at the auditory brainstem after exposing rats to a pure tone or a narrow-range frequency modulated (FM) sound for various durations (10, 30 or 90 min). Nuclear sizes of Fos-stains at the cochlear nucleus (CN) and inferior colliculus (IC) were estimated under microscope. Tone stimulation at the given frequency (9 kHz) induced Fos-expression at locations consistent with the tonotopic maps, and no clear difference in the spatial distribution of the Fos-stains was observed across stimuli. In general, more Fos-stains appeared after longer stimulations and most notably cell nuclei labeled with Fos-immunoreactivity were statistically larger in size under longer pure tone and FM tone stimulations. Findings suggest that Fos-expression mechanisms are related to the selective response of different subpopulations of neurons to sounds of different time-varying properties. Results support that the time-variance of acoustic stimulation should be considered in the interpretation of Fos-expression findings.


Assuntos
Percepção Auditiva/fisiologia , Núcleo Celular/metabolismo , Núcleo Coclear/metabolismo , Colículos Inferiores/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estimulação Acústica , Animais , Vias Auditivas/citologia , Vias Auditivas/metabolismo , Mapeamento Encefálico , Núcleo Celular/ultraestrutura , Tamanho Celular , Núcleo Coclear/citologia , Imuno-Histoquímica , Colículos Inferiores/citologia , Neurônios/citologia , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Ratos Sprague-Dawley , Tempo de Reação/fisiologia , Fatores de Tempo
3.
Eur J Neurosci ; 27(7): 1634-46, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18371080

RESUMO

WW domain-containing oxidoreductase (named WWOX, FOR or WOX1) is a pro-apoptotic protein and tumor suppressor. Animals treated with dopaminergic neurotoxin 1-methyl-4-phenyl-pyridinium (MPP+) develop Parkinson's disease (PD)-like symptoms. Here we investigated whether WOX1 is involved in MPP+-induced neurodegeneration. Upon insult with MPP+ in rat brains, WOX1 protein was upregulated and phosphorylated at Tyr33 (or activated) in the injured neurons in the striatum and cortex ipsilaterally to intoxication, as determined by immunohistochemistry and Western blotting. Also, WOX1 was present in the condensed nuclei and damaged mitochondria of degenerative neurons, as revealed by transmission immunoelectron microscopy. Time-lapse microscopy revealed that MPP+ induced membrane blebbing and shrinkage of neuroblastoma SK-N-SH cells. Dominant-negative WOX1, a potent inhibitor of Tyr33 phosphorylation, abolished this event, indicating a critical role of the phosphorylation in apoptosis. c-Jun N-terminal kinase (JNK1) is known to bind and counteract the apoptotic function of WOX1. Suppression of JNK1 function by a dominant-negative spontaneously induced WOX1 activation. WOX1 physically interacted with JNK1 in SK-N-SH cells and rat brain extracts. MPP+ rapidly increased the binding, followed by dissociation, which is probably needed for WOX1 to exert apoptosis. We synthesized a short Tyr33-phosphorylated WOX1 peptide (11 amino acid residues). Interestingly, this peptide blocked MPP+-induced neuronal death in the rat brains, whereas non-phospho-WOX1 peptide had no effect. Together, activated WOX1 plays an essential role in the MPP+-induced neuronal death. Our synthetic phospho-WOX1 peptide prevents neuronal death, suggestive of its therapeutic potential in mitigating the symptoms of PD.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Apoptose/fisiologia , Neurônios/enzimologia , Oxirredutases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Tirosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Humanos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Oxirredutases/genética , Doença de Parkinson/enzimologia , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Supressoras de Tumor/genética , Tirosina/genética , Oxidorredutase com Domínios WW
4.
Oncotarget ; 8(12): 19137-19155, 2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-27845895

RESUMO

Malignant cancer cells frequently secrete significant amounts of transforming growth factor beta (TGF-ß), hyaluronan (HA) and hyaluronidases to facilitate metastasizing to target organs. In a non-canonical signaling, TGF-ß binds membrane hyaluronidase Hyal-2 for recruiting tumor suppressors WWOX and Smad4, and the resulting Hyal-2/WWOX/Smad4 complex is accumulated in the nucleus to enhance SMAD-promoter dependent transcriptional activity. Yeast two-hybrid analysis showed that WWOX acts as a bridge to bind both Hyal-2 and Smad4. When WWOX-expressing cells were stimulated with high molecular weight HA, an increased formation of endogenous Hyal-2/WWOX/Smad4 complex occurred rapidly, followed by relocating to the nuclei in 20-40 min. In WWOX-deficient cells, HA failed to induce Smad2/3/4 relocation to the nucleus. To prove the signaling event, we designed a real time tri-molecular FRET analysis and revealed that HA induces the signaling pathway from ectopic Smad4 to WWOX and finally to p53, as well as from Smad4 to Hyal-2 and then to WWOX. An increased binding of the Smad4/Hyal-2/WWOX complex occurs with time in the nucleus that leads to bubbling cell death. In contrast, HA increases the binding of Smad4/WWOX/p53, which causes membrane blebbing but without cell death. In traumatic brain injury-induced neuronal death, the Hyal-2/WWOX complex was accumulated in the apoptotic nuclei of neurons in the rat brains in 24 hr post injury, as determined by immunoelectron microscopy. Together, HA activates the Hyal-2/WWOX/Smad4 signaling and causes bubbling cell death when the signaling complex is overexpressed.


Assuntos
Morte Celular/fisiologia , Ácido Hialurônico/metabolismo , Neoplasias/patologia , Oxirredutases/metabolismo , Transdução de Sinais/fisiologia , Proteína Smad4/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Transferência Ressonante de Energia de Fluorescência , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Neoplasias/metabolismo , Ratos , Técnicas do Sistema de Duplo-Híbrido , Oxidorredutase com Domínios WW
5.
Clin Cancer Res ; 11(16): 5769-77, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16115915

RESUMO

PURPOSE: We investigated the role of candidate tumor suppressor and proapoptotic WOX1 (also named WWOX, FOR, or WWOXv1) in UVB-induced apoptosis and formation of cutaneous squamous cell carcinomas (SCC). EXPERIMENTAL DESIGN: Expression of WOX1 and family proteins (WWOX) in human primary cutaneous SCCs was examined by immunohistochemistry, in situ hybridization, and reverse transcription-PCR. UVB irradiation-induced WOX1 activation (Tyr33 phosphorylation and nuclear translocation), apoptosis, and cutaneous SCC formation were examined both in vitro and in vivo. RESULTS: Up-regulation of human WOX1, isoform WOX2, and Tyr33 phosphorylation occurred during normal keratinocyte differentiation before cornification and death. Interestingly, significant reduction of these proteins and Tyr33 phosphorylation was observed in nonmetastatic and metastatic cutaneous SCCs (P < 0.001), but without down-regulation of WWOX mRNA (P > 0.05 versus normal controls), indicating a translational blockade of WWOX mRNA to protein. During acute exposure of hairless mice to UVB, WOX1 was up-regulated and activated in epidermal cells in 24 hours. In parallel with the clinical findings in humans, chronic UVB-treated mice developed cutaneous SCCs in 3 months, with significant reduction of WOX1 and Tyr33 phosphorylation and, again, without down-regulation of WWOX mRNA. Human SCC-25 and HaCaT cells were transfected with small interfering RNA-targeting WOX1 and shown to resist UVB-induced WOX1 expression, activation, and apoptosis. CONCLUSIONS: WOX1 is essential for UVB-induced apoptosis and likely to be involved in the terminal differentiation of normal keratinocytes. During UVB-induced cutaneous SCC, epidermal cells have apparently prevented the apoptotic pressure from overexpressed WOX1 by shutting down the translation machinery for WWOX mRNA.


Assuntos
Apoptose/efeitos da radiação , Carcinoma de Células Escamosas/genética , Oxirredutases/genética , Biossíntese de Proteínas/genética , Neoplasias Cutâneas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Imuno-Histoquímica , Hibridização In Situ , Queratinócitos/citologia , Queratinócitos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Oxirredutases/metabolismo , Fosforilação/efeitos da radiação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/metabolismo , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor , Tirosina/metabolismo , Raios Ultravioleta , Oxidorredutase com Domínios WW
6.
Biochem Pharmacol ; 66(8): 1347-54, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14555208

RESUMO

Human WWOX gene encodes a putative tumor suppressor WW domain-containing oxidoreductase WOX1 (also known as WWOX or FOR). A high frequency of loss of heterozygosity (LOH) of this gene has been shown in prostate, lung, breast and other cancers. In addition, numerous aberrant WWOX mRNA transcripts have been found in cancer cells. WOX1 is a proapoptotic protein. In response to stress or apoptotic stimuli, WOX1 became phosphorylated at Tyr33, which enabled its complex formation with activated p53 and JNK1. The p53/WOX1 complex translocated to the mitochondria and further to the nuclei to mediate apoptosis. WOX1 mutants, which were inactivated for nuclear translocation or Tyr33 phosphorylation, failed to induce apoptosis, indicating that activation of WOX1 via Tyr33 phosphorylation, followed by nuclear translocation, is essential for inducing cell death. WOX1 induced apoptosis synergistically with p53. In contrast, transiently activated JNK1 induced anti-apoptotic response, and this protective activity inhibited WOX1-induced apoptosis. Taken together, WOX1 is involved in stress and apoptotic responses, and is likely to regulate the activation of both p53 and JNK1.


Assuntos
Apoptose/fisiologia , Oxirredutases/metabolismo , Estresse Fisiológico/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Transporte Biológico , Núcleo Celular/metabolismo , Insuficiência Cardíaca , Humanos , Mitocôndrias/metabolismo , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Fator de Necrose Tumoral alfa/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Oxidorredutase com Domínios WW
7.
Neurochem Int ; 40(2): 169-79, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11738483

RESUMO

While there is growing evidence that Bcl-2 proto-oncogene and beta-amyloid precursor proteins (APP) are neuroprotective in function, our recent studies have demonstrated that Bcl-2 and APP may be co-expressed and co-regulated in retinal neurons or glia under normal or experimental conditions. Whether Bcl-2 and APP are functionally coupled in other neuronal systems is not clear. This issue was investigated further in the present experiments by examining the expression pattern of two molecules after unilateral intrastriatal injection of 1-methyl-4-phenyl-pyridinium (MPP(+)), a neurotoxic metabolite that selectively damages dopaminergic neurons. One hour to 2 months after MPP(+) injection into rat striatum, a significant increase in Bcl-2 expression was observed in distinct populations of neurons, astrocyte-like and OX-42-positive cells not only in traumatic regions but also in remote areas including the ipsilateral cortex and substantia nigra (SN). No detectable change was observed in the striatum, cortex or SN on the contralateral side of the brain. The immunoreactive pattern and time-dependent APP increase was similar to that of Bcl-2 in the severely injured striatum and cortex. However, an up-regulation of Bcl-2 expression, but not APP, appears in dopaminergic neurons in the ipsilateral SN pars compacta where there was retrograde degeneration. In contrast, APP immunoreactivity was decreased in the hippocampus following intrastriatal injury, whereas, no alteration in Bcl-2 expression was detected. The differential changes in Bcl-2 and APP expression in nigral neurons and some other brain tissues suggest that these proteins may not be co-regulated by a common mechanism, at least in certain neuronal pathways.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Precursor de Proteína beta-Amiloide/genética , Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes bcl-2/efeitos dos fármacos , Neurônios/fisiologia , Substância Negra/fisiologia , 1-Metil-4-fenilpiridínio/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Córtex Cerebral/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Lateralidade Funcional , Regulação da Expressão Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Masculino , Microinjeções , Neurônios/classificação , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos
8.
Oncotarget ; 5(23): 11792-9, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25537520

RESUMO

The human and mouse WWOX/Wwox gene encodes a candidate tumor suppressor WW domain-containing oxidoreductase protein. This gene is located on a common fragile site FRA16D. WWOX participates in a variety of cellular events and acts as a transducer in the many signal pathways, including TNF, chemotherapeutic drugs, UV irradiation, Wnt, TGF-ß, C1q, Hyal-2, sex steroid hormones, and others. While transiently overexpressed WWOX restricts relocation of transcription factors to the nucleus for suppressing cancer survival, physiological relevance of this regard in vivo has not been confirmed. Unlike many tumor suppressor genes, mutation of WWOX is rare, raising a question whether WWOX is a driver for cancer initiation. WWOX/Wwox was initially shown to play a crucial role in neural development and in the pathogenesis of Alzheimer's disease and neuronal injury. Later on, WWOX/Wwox was shown to participate in the development of epilepsy, mental retardation, and brain developmental defects in mice, rats and humans. Up to date, most of the research and review articles have focused on the involvement of WWOX in cancer. Here, we review the role of WWOX in neural injury and neurological diseases, and provide perspectives for the WWOX-regulated neurodegeneration.


Assuntos
Doenças do Sistema Nervoso , Oxirredutases , Proteínas Supressoras de Tumor , Animais , Humanos , Camundongos , Oxidorredutase com Domínios WW
9.
Anal Cell Pathol (Amst) ; 36(5-6): 133-47, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24503545

RESUMO

BACKGROUND AND OBJECTIVES: We aimed to evaluate the expression levels of the tumor suppressor WOX1 in nervous system tumors and its co-expression with p53 and neurofibromatosis type 2/merlin (NF2) tumor suppressor gene products. METHODS: Immunohistochemistry, western blotting and in situ hybridization were used for WOX1 protein and WWOX mRNA expression. Immunofluorescence and electron microscopical immunohistochemistry were performed for colocalization of gene products. RESULTS: WOX1 expression is low in normal cortical neurons, mainly on the axon fibers, whereas there is moderate to high immunoreactivity in the cytosol and nuclei of certain tumor cells. In the microcystic (WHO grade I) and malignant (WHO grade III) meningiomas, WOX1 expression is intense, but various in transitional (WHO grade I) and atypical (WHO grade II) subtypes. WOX1 levels are moderate to high in the menigiotheliomatous area, but relatively low in the fibroblastic area. WOX1 and NF2/merlin, but not p53, colocalized in certain tumor cells, primarily at the borders of nuclei. Schwannoma and astrocytoma specimens stained moderately to strongly positive for the WOX1 protein. Interestingly, the expression of WOX1, NF2/merlin and mutant p53 is intense in high grade glioblastoma, but WOX1 expression is low in metastatic carcinoma or adenocarcinoma. CONCLUSIONS: The expression of WOX1 on different types of nervous system tumors, including primary and metastatic tumors, is differential.


Assuntos
Neoplasias do Sistema Nervoso/metabolismo , Oxirredutases/química , Oxirredutases/metabolismo , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/metabolismo , Criança , Feminino , Humanos , Masculino , Meningioma/metabolismo , Pessoa de Meia-Idade , Oxidorredutase com Domínios WW , Adulto Jovem
10.
Invest Ophthalmol Vis Sci ; 52(7): 4214-22, 2011 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-21345998

RESUMO

PURPOSE: Benzyl alcohol (BA) is the preservative in triamcinolone acetonide (TA) suspensions, which are used in treating vitreoretinal diseases and during surgery. This paper investigates the molecular mechanisms and signaling pathways underlying BA toxicity in human retinal pigment epithelial (RPE) cells. METHODS: Cultured human RPE cells from the ARPE-19 cell line were exposed to culture medium alone (control) or with BA (0.0225, 0.225, 0.9, 3, or 9 mg/mL) for up to 6 hours. BA toxicity was assessed by TUNEL assay, propidium iodide/annexin V-FITC staining and flow cytometry, caspase activation assay, caspase and apoptosis inhibition assays, mitochondrial transmembrane potential by rhodamine staining and flow cytometry, reactive oxygen species by chemiluminescence, and apoptosis-inducing factor staining. RESULTS: BA caused RPE cell death not only by necrosis but also by apoptosis, evidenced by exposure to 9 mg/mL BA for 6 hours leading to 19.0% early apoptotic cells and 64.2% apoptotic necrotic cells. Apoptotic signaling involved the immediate production of reactive oxygen species, activation of caspase-8, impairment of the mitochondrial transmembrane potential, and further activation of caspase-9 and -3. In addition, BA induced translocation of apoptosis-inducing factor into the nucleus, indicating caspase-independent apoptosis. CONCLUSIONS: BA leads to necrosis of RPE cells and triggers mitochondrial apoptosis through both caspase-dependent and - independent pathways. Extreme caution is suggested in the intraocular use of TA suspensions and meticulous evaluation before adoption of BA as a preservative in the future development of ophthalmic formulations.


Assuntos
Álcool Benzílico/toxicidade , Glucocorticoides , Conservantes Farmacêuticos/toxicidade , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia , Triancinolona Acetonida , Apoptose , Fator de Indução de Apoptose/metabolismo , Álcool Benzílico/administração & dosagem , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Esquema de Medicação , Ativação Enzimática , Humanos , Potenciais da Membrana , Mitocôndrias , Necrose , Conservantes Farmacêuticos/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais , Coloração e Rotulagem , Fatores de Tempo , Distribuição Tecidual/efeitos dos fármacos
11.
Neurosci Lett ; 463(2): 145-9, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19646507

RESUMO

Prolonged sound exposure produces functional changes in the auditory neurons. It remains unclear whether such changes are detectable with morphometric measures like cell size. Here, after exposing juvenile rats (starting on week-4) to a monotone for 7 days, we measured the size of their cortical neurons. Neuronal profiles (nuclei and perikarya) in deep layers of the primary auditory cortex were digitized and measured on photomicrographs taken from 7 microm-thick histological sections stained with toluidine blue. To facilitate digitizing cell profiles, we used an image-analysis software that contains a confocal-like image-merging function to sharpen the edges. After sound exposure, both nuclei and perikarya expanded by about 1/3 in volume compared with controls (p<0.0001, Student's t-test). Such changes were not found in the visual cortex. Results showed that prolonged sound exposure increased the size of auditory neurons. Such activity-driven cell enlargement can be used as a simple measure to find other plastic changes in the brain.


Assuntos
Córtex Auditivo/citologia , Neurônios/citologia , Som , Animais , Tamanho Celular , Masculino , Ratos , Ratos Sprague-Dawley
12.
J Biol Chem ; 284(23): 16049-59, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19366691

RESUMO

Transforming growth factor beta (TGF-beta) initiates multiple signal pathways and activates many downstream kinases. Here, we determined that TGF-beta1 bound cell surface hyaluronidase Hyal-2 on microvilli in type II TGF-beta receptor-deficient HCT116 cells, as determined by immunoelectron microscopy. This binding resulted in recruitment of proapoptotic WOX1 (also named WWOX or FOR) and formation of Hyal-2.WOX1 complexes for relocation to the nuclei. TGF-beta1 strengthened the binding of the catalytic domain of Hyal-2 with the N-terminal Tyr-33-phosphorylated WW domain of WOX1, as determined by time lapse fluorescence resonance energy transfer analysis in live cells, co-immunoprecipitation, and yeast two-hybrid domain/domain mapping. In promoter activation assay, ectopic WOX1 or Hyal-2 alone increased the promoter activity driven by Smad. In combination, WOX1 and Hyal-2 dramatically enhanced the promoter activation (8-9-fold increases), which subsequently led to cell death (>95% of promoter-activated cells). TGF-beta1 supports L929 fibroblast growth. In contrast, transiently overexpressed WOX1 and Hyal-2 sensitized L929 to TGF-beta1-induced apoptosis. Together, TGF-beta1 invokes a novel signaling by engaging cell surface Hyal-2 and recruiting WOX1 for regulating the activation of Smad-driven promoter, thereby controlling cell growth and death.


Assuntos
Moléculas de Adesão Celular/fisiologia , Membrana Celular/fisiologia , Hialuronoglucosaminidase/fisiologia , Oxirredutases/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Moléculas de Adesão Celular/genética , Neoplasias Colorretais , Primers do DNA , Transferência Ressonante de Energia de Fluorescência , Proteínas Ligadas por GPI , Regulação da Expressão Gênica , Células HCT116 , Humanos , Hialuronoglucosaminidase/genética , Células L , Camundongos , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/deficiência , Ratos , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Oxidorredutase com Domínios WW
13.
PLoS One ; 4(11): e7820, 2009 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19918364

RESUMO

BACKGROUND: Tumor suppressor WOX1 (also named WWOX or FOR) is known to participate in neuronal apoptosis in vivo. Here, we investigated the functional role of WOX1 and transcription factors in the delayed loss of axotomized neurons in dorsal root ganglia (DRG) in rats. METHODOLOGY/PRINCIPAL FINDINGS: Sciatic nerve transection in rats rapidly induced JNK1 activation and upregulation of mRNA and protein expression of WOX1 in the injured DRG neurons in 30 min. Accumulation of p-WOX1, p-JNK1, p-CREB, p-c-Jun, NF-kappaB and ATF3 in the nuclei of injured neurons took place within hours or the first week of injury. At the second month, dramatic nuclear accumulation of WOX1 with CREB (>65% neurons) and NF-kappaB (40-65%) occurred essentially in small DRG neurons, followed by apoptosis at later months. WOX1 physically interacted with CREB most strongly in the nuclei as determined by FRET analysis. Immunoelectron microscopy revealed the complex formation of p-WOX1 with p-CREB and p-c-Jun in vivo. WOX1 blocked the prosurvival CREB-, CRE-, and AP-1-mediated promoter activation in vitro. In contrast, WOX1 enhanced promoter activation governed by c-Jun, Elk-1 and NF-kappaB. WOX1 directly activated NF-kappaB-regulated promoter via its WW domains. Smad4 and p53 were not involved in the delayed loss of small DRG neurons. CONCLUSIONS/SIGNIFICANCE: Rapid activation of JNK1 and WOX1 during the acute phase of injury is critical in determining neuronal survival or death, as both proteins functionally antagonize. In the chronic phase, concurrent activation of WOX1, CREB, and NF-kappaB occurs in small neurons just prior to apoptosis. Likely in vivo interactions are: 1) WOX1 inhibits the neuroprotective CREB, which leads to eventual neuronal death, and 2) WOX1 enhances NF-kappaB promoter activation (which turns to be proapoptotic). Evidently, WOX1 is the potential target for drug intervention in mitigating symptoms associated with neuronal injury.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Gânglios Espinais/metabolismo , NF-kappa B/metabolismo , Neurônios/metabolismo , Oxirredutases/metabolismo , Nervo Isquiático/cirurgia , Animais , Apoptose , Ativação Enzimática , Masculino , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
14.
J Pineal Res ; 42(3): 297-309, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17349029

RESUMO

We have previously shown that exogenous melatonin improves the preservation of the blood-brain barrier (BBB) and neurovascular unit following cerebral ischemia-reperfusion. Recent evidence indicates that postischemic microglial activation exaggerates the damage to the BBB. Herein, we explored whether melatonin mitigates the cellular inflammatory response after transient focal cerebral ischemia for 90 min in rats. Melatonin (5 mg/kg) or vehicle was given intravenously at reperfusion onset. Immunohistochemistry and flow cytometric analysis were used to evaluate the cellular inflammatory response at 48 hr after reperfusion. Relative to controls, melatonin-treated animals did not have significantly changed systemic cellular inflammatory responses in the bloodstream (P > 0.05). Melatonin, however, significantly decreased the cellular inflammatory response by 41% (P < 0.001) in the ischemic hemisphere. Specifically, melatonin effectively decreased the extent of neutrophil emigration (Ly6G-positive/CD45-positive) and macrophage/activated microglial infiltration (CD11b-positive/CD45-positive) by 51% (P < 0.01) and 66% (P < 0.01), respectively, but did not significantly alter the population composition of T lymphocyte (CD3-positive/CD45-positive; P > 0.05). This melatonin-mediated decrease in the cellular inflammatory response was accompanied by both reduced brain infarction and improved neurobehavioral outcome by 43% (P < 0.001) and 50% (P < 0.001), respectively. Thus, intravenous administration of melatonin upon reperfusion effectively decreased the emigration of circulatory neutrophils and macrophages/monocytes into the injured brain and inhibited focal microglial activation following cerebral ischemia-reperfusion. The finding demonstrates melatonin's inhibitory ability against the cellular inflammatory response after cerebral ischemia-reperfusion, and further supports its pleuripotent neuroprotective actions suited either as a monotherapy or an add-on to the thrombolytic therapy for ischemic stroke patients.


Assuntos
Inflamação/tratamento farmacológico , Ataque Isquêmico Transitório/tratamento farmacológico , Leucócitos/efeitos dos fármacos , Melatonina/administração & dosagem , Animais , Injeções Intravenosas , Masculino , Microglia/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Reperfusão
15.
J Pineal Res ; 41(2): 175-82, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16879324

RESUMO

We have recently shown that melatonin decreases the late (24 hr) increase in blood-brain barrier (BBB) permeability and the risk of tissue plasminogen activator-induced hemorrhagic transformation following ischemic stroke in mice. In the study, we further explored whether melatonin would reduce postischemic neurovascular oxidative/nitrosative damage and, therefore, improve preservation of the early increase in the BBB permeability at 4 hr after transient focal cerebral ischemia for 60 min in mice. Melatonin (5 mg/kg) or vehicle was given intraperitoneally at the beginning of reperfusion. Hydroethidine (HEt) in situ detection and immunohistochemistry for nitrotyrosine were used to evaluate postischemic accumulation in reactive oxygen and nitrogen species, respectively, in the ischemic neurovascular unit. BBB permeability was evaluated by spectrophotometric and microscopic quantitation of Evans Blue leakage. Relative to controls, melatonin-treated animals not only had a significantly reduced superoxide accumulation in neurovascular units in boundary zones of infarction, by reducing 35% and 54% cytosolic oxidized HEt in intensity and cell-expressing percentage, respectively (P < 0.001), but also exhibited a reduction in nitrotyrosine by 52% (P < 0.01). Additionally, melatonin-treated animals had significantly reduced early postischemic disruption in the BBB permeability by 53% (P < 0.001). Thus, melatonin reduced postischemic oxidative/nitrosative damage to the ischemic neurovascular units and improved the preservation of BBB permeability at an early phase following transient focal cerebral ischemia in mice. The findings further highlight the ability of melatonin in anatomical and functional preservation for the ischemic neurovascular units and its relevant potential in the treatment of ischemic stroke.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Infarto da Artéria Cerebral Média/fisiopatologia , Ataque Isquêmico Transitório/fisiopatologia , Melatonina/farmacologia , Animais , Encéfalo/irrigação sanguínea , Azul Evans/farmacocinética , Ataque Isquêmico Transitório/tratamento farmacológico , Fluxometria por Laser-Doppler , Masculino , Melatonina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/fisiopatologia
16.
J Pineal Res ; 38(1): 42-52, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15617536

RESUMO

We have previously shown that melatonin reduces infarct volumes and enhances neurobehavioral and electrophysiological recoveries following transient middle cerebral artery (MCA) occlusion in rats. In the study, we examined whether melatonin would display neuroprotection against neuronal, axonal and oligodendrocyte pathology after 24 hr of reperfusion following 1 hr of MCA occlusion in mice. Melatonin (5 mg/kg) or vehicle was given intraperitoneally at the commencement of reperfusion. Neurological deficits were assessed 24 hr after ischemia. Gray matter damage was evaluated by quantitative histopathology. Axonal damage was determined with amyloid precursor protein and microtubule-associated protein tau-1 immunohistochemistry to identify postischemic disrupted axonal flow and oligodendrocyte pathology, respectively. Oxidative damage was assessed by 8-hydroxy-2'-deoxyguanosine (8-OHdG) and 4-hydroxynonenal (4-HNE) immunohistochemistry. Relative to controls, melatonin-treated animals not only had a significantly reduced volume of gray matter infarction by 42% (P<0.001), but also exhibited a decreased score of axonal damage by 42% (P<0.001) and a reduction in the volume of oligodendrocyte pathology by 58% (P<0.005). Melatonin-treated animals also had significantly reduced immunopositive reactions for 8-OHdG and 4-HNE by 53% (P<0.001) and 49% (P<0.001), respectively. In addition, melatonin improved sensory and motor neurobehavioral outcomes by 47 and 30%, respectively (P<0.01). Thus, delayed (1 hr) treatment with melatonin reduced both gray and white matter damage and improved neurobehavioral outcomes following transient focal cerebral ischemia in mice. The finding of reduced oxidative damage observed with melatonin suggests that its major mechanisms of action are mediated through its antioxidant and radical scavenging activity.


Assuntos
Isquemia Encefálica/prevenção & controle , Encéfalo/patologia , Melatonina/farmacologia , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , DNA/metabolismo , Hidroxilação , Peroxidação de Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo
17.
J Med Virol ; 70(1): 109-18, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12629651

RESUMO

The cell tropism of enterovirus 71 (Enteroviridae) in neuronal, glial and laryngeal cells. The 4643 strain, an enterovirus 71 isolate from a patient in Taiwan, was used to infect three human cell lines representing neuronal cells (SK-N-SH, neuroblastoma), glial cells (U373MG, glioblastoma), and laryngeal cells (HEp-2, larynx epidermoid carcinoma). Immunofluorescent staining and transmission electron microscopy (TEM) were used to detect mature enterovirus 71 4643 virions in these cell lines. The three cell lines were also compared for presence of virus-mediated cytopathic effect (CPE), synthesis of infected cell-specific proteins, viral (-) RNA, and virus replication rate. Virus particles were detected by TEM, and viral replication increased over time, indicating the existence and release of mature viruses from all three infected cell lines. The most severe CPE and the highest viral replication rate were observed in the SK-N-SH cells. Further screening of the infected cell lines by microarray analysis revealed that the neuron growth factor receptor (NGFR) gene was uniquely upregulated in infected SK-N-SH cells, implying that the receptor encoded by this gene may be involved in cell tropism. The data show that neurons are vulnerable to enterovirus 71 4643 infection and are consistent with the clinical observation that enterovirus 71 4643 targets mainly neuronal cells but is also found in many organs in conjunction with an inflammatory reaction.


Assuntos
Enterovirus/fisiologia , Neurônios/virologia , Replicação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Efeito Citopatogênico Viral , Enterovirus/crescimento & desenvolvimento , Regulação Viral da Expressão Gênica , Humanos , Microscopia Eletrônica , Neuroglia , RNA Viral/biossíntese , Células Vero , Proteínas Virais/biossíntese , Cultura de Vírus
18.
J Pineal Res ; 32(4): 262-9, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11982797

RESUMO

In this study we selected a rat model of Parkinson's disease (PD) by using intrastriatal infusion of the 1-methyl-4-phenyl-pyridinium ion (MPP+) to investigate the neuroprotective action of melatonin and its inhibitory activity on MPP+-impaired glutathione (GSH) system in the nigrostriatal system. Results show that MPP+ caused not only a severe neuronal injury in the striatum and in the ipsilateral substantia nigra (SN), but it also induced a significant decrease in GSH levels and an increase in the GSSG/GSH ratio 3 days after intrastriatal MPP+ infusion. Intraperitoneal co-administration of melatonin (10 mg/kg, five times) significantly attenuated MPP+-induced nigrostriatal neurotoxicity and GSH impairment. Depletion of cytosolic GSH by L-buthionine sulfoximine (BSO) did not cause neuronal damage by itself. It, however, when co-administrated with MPP+, potentiated the GSH reduction in the striatum, without aggravating nigrostriatal neurodegeneration induced by MPP+. Moreover, the MPP+-caused neuronal damage was positively correlated with a rising ratio of GSSG/GSH, but not with a drop of GSH. These results suggest that the MPP+-triggered oxidative stress may play a more important role than the loss of the antioxidant GSH in determining neuronal injury. Interestingly, the neuronal damage and oxidative stress elicited by co-treatment of BSO with MPP+ were effectively reduced by melatonin. Our results hence provide direct evidence showing that melatonin attenuates MPP+-induced nigrostriatal dopaminergic injury by its ability to impede the increase of GSSG/GSH ratio; therefore melatonin may have therapeutic implications in PD.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Corpo Estriado/efeitos dos fármacos , Glutationa/fisiologia , Melatonina/farmacologia , Substância Negra/efeitos dos fármacos , Animais , Corpo Estriado/metabolismo , Dopamina/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Ratos , Ratos Wistar , Substância Negra/enzimologia , Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
19.
Neurobiol Dis ; 17(3): 367-77, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15571973

RESUMO

The objective of this study was to search for brain-specific binding proteins that participated in Abeta aggregation. Immunoprecipitation of Abeta in Alzheimer's brain homogenate revealed a major co-precipitating 16-kDa protein band, which was identified through mass spectrometry as hemoglobin (Hb) alpha and beta chains. Hemoglobin was distributed in Alzheimer's disease (AD) patients in a brain region-dependent manner, with the highest levels in the hippocampus and parietal gray (PG) matter, followed by parietal white matter (PW), and the lowest in cerebellum (Cb). AD parietal gray and white matters exhibited higher Hb levels than those in the nondemented (ND) group. Likewise, RT-PCR revealed that the Hb mRNA levels in AD inferior temporal gyri were higher than those of ND subjects. Furthermore, Hb was shown to promote Abeta oligomer formation. Immunohistochemical studies indicated that Hb was localized within the cytosol of pyramidal neurons in the hippocampus, suggesting a potential source of intracerebral Hb. Finally, double immunofluorescent assay confirmed the co-localization of Hb with senile plaques (SP) and cerebral amyloid angiopathy (CAA). We propose that an elevation in brain Hb via circulation leakage or perturbations of Hb gene regulation may participate in AD pathogenesis.


Assuntos
Doença de Alzheimer/patologia , Amiloide/metabolismo , Encéfalo/patologia , Hemoglobinas/fisiologia , Neurônios/patologia , Idoso , Idoso de 80 Anos ou mais , Primers do DNA , Feminino , Hemoglobinas/análise , Hemoglobinas/química , Humanos , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Estrutura Quaternária de Proteína , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA