Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-36835121

RESUMO

The pathogenesis of Parkinson's disease (PD) is very complex and still needs further exploration. Leucine-rich repeat kinase 2 (LRRK2) is associated with familial PD in mutant forms and sporadic PD in the wild-type form. Abnormal iron accumulation is found in the substantia nigra of PD patients, but its exact effects are not very clear. Here, we show that iron dextran exacerbates the neurological deficit and loss of dopaminergic neurons in 6-OHDA lesioned rats. 6-OHDA and ferric ammonium citrate (FAC) significantly increase the activity of LRRK2 as reflected by the phosphorylation of LRRK2, at S935 and S1292 sites. 6-OHDA-induced LRRK2 phosphorylation is attenuated by the iron chelator deferoxamine, especially at the S1292 site. 6-OHDA and FAC markedly induce the expression of pro-apoptotic molecules and the production of ROS by activating LRRK2. Furthermore, G2019S-LRRK2 with high kinase activity showed the strongest absorptive capacity for ferrous iron and the highest intracellular iron content among WT-LRRK2, G2019S-LRRK2, and kinase-inactive D2017A-LRRK2 groups. Taken together, our results demonstrate that iron promotes the activation of LRRK2, and active LRRK2 accelerates ferrous iron uptake, suggesting that there exists an interplay between iron and LRRK2 in dopaminergic neurons, providing a new perspective to uncover the underlying mechanisms of PD occurrence.


Assuntos
Ferro , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Doença de Parkinson , Animais , Ratos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Ferro/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Mutação , Oxidopamina/farmacologia , Oxidopamina/toxicidade , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Substância Negra/metabolismo , Substância Negra/patologia , Modelos Animais de Doenças
2.
Asian J Androl ; 25(3): 356-360, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36254889

RESUMO

Enhanced recovery after surgery (ERAS) measures have not been systematically applied in transurethral surgery for benign prostatic hyperplasia (BPH). This study was performed on patients with BPH who required surgical intervention. From July 2019 to June 2020, the ERAS program was applied to 248 patients, and the conventional program was applied to 238 patients. After 1 year of follow-up, the differences between the ERAS group and the conventional group were evaluated. The ERAS group had a shorter time of urinary catheterization compared with the conventional group (mean ± standard deviation [s.d.]: 1.0 ± 0.4 days vs 2.7 ± 0.8 days, P < 0.01), and the pain (mean ± s.d.) was significantly reduced through postoperative hospitalization days (PODs) 0-2 (POD 0: 1.7 ± 0.8 vs 2.4 ± 1.0, P < 0.01; POD 1: 1.6 ± 0.9 vs 3.5 ± 1.3, P < 0.01; POD 2: 1.2 ± 0.7 vs 3.0 ± 1.3, P < 0.01). No statistically significant difference was found in the rate of postoperative complications, such as postoperative bleeding (P = 0.79), urinary retention (P = 0.40), fever (P = 0.55), and readmission (P = 0.71). The hospitalization cost of the ERAS group was similar to that of the conventional group (mean ± s.d.: 16 927.8 ± 5808.1 Chinese Yuan [CNY] vs 17 044.1 ± 5830.7 CNY, P =0.85). The International Prostate Symptom Scores (IPSS) and quality of life (QoL) scores in the two groups were also similar when compared at 1 month, 3 months, 6 months, and 12 months after discharge. The ERAS program we conducted was safe, repeatable, and efficient. In conclusion, patients undergoing the ERAS program experienced less postoperative stress than those undergoing the conventional program.


Assuntos
Recuperação Pós-Cirúrgica Melhorada , Hiperplasia Prostática , Ressecção Transuretral da Próstata , Masculino , Humanos , Hiperplasia Prostática/complicações , Qualidade de Vida , Ressecção Transuretral da Próstata/efeitos adversos , Resultado do Tratamento
3.
Front Mol Neurosci ; 15: 1045778, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36545123

RESUMO

The central nervous system is the most important and difficult to study system in the human body and is known for its complex functions, components, and mechanisms. Neurons are the basic cellular units realizing neural functions. In neurons, vesicles are one of the critical pathways for intracellular material transport, linking information exchanges inside and outside cells. The axon is a vital part of neuron since electrical and molecular signals must be conducted through axons. Here, we describe and explore the formation, trafficking, and sorting of cellular vesicles within axons, as well as related-diseases and practical implications. Furthermore, with deepening of understanding and the development of new approaches, accumulating evidence proves that besides signal transmission between synapses, the material exchange and vesicular transmission between axons and extracellular environment are involved in physiological processes, and consequently to neural pathology. Recent studies have also paid attention to axonal vesicles and their physiological roles and pathological effects on axons themselves. Therefore, this review mainly focuses on these two key nodes to explain the role of intracellular vesicles and extracellular vesicles migrated from cells on axons and neurons, providing innovative strategy for future researches.

4.
Zhongguo Zhong Yao Za Zhi ; 44(22): 4800-4805, 2019 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-31872585

RESUMO

Due to the increasing incidence of central nervous system diseases,especially the increasing incidence and mortality of stroke,brain-targeted drug delivery has attached more and more attention. Nasal administration,as one of the ways of brain-targeted administration,can effectively make the drug delivered to the brain in a targeted way after by passing the blood-brain barrier,providing a new idea for the treatment of central nervous system diseases. Therefore,it is a promising administration way. In recent years,the treatment of encephalopathy by nasal administration of traditional Chinese medicine has become a hot topic in the research of traditional Chinese medicine. Ischemic stroke is one of the most important diseases endangering human health. Nasal administration has a history of thousands of years in treatment of stroke. Modern medical research has proved that there is a subtle connection between the nasal cavity and the brain,and the complex and ingenious structure of the nasal cavity provides the possibility for drugs delivery to the brain through the nose. Drug administration through nasal cavity has obvious advantages in treatment of central nervous system diseases represented by ischemic stroke. Nasal administration is characterized by non-invasion,low infection,rapid absorption and brain targeting. The author will expound the theoretical basis of brain targeting of nasal administration from the aspects of anatomy and physiology,and summarize the transport pathway of drugs through the nose into the brain,the in vitro and in vivo experimental research basis of the " nose-brain"pathway,and the clinical nasal administration of traditional Chinese medicine to prevent cerebral ischemia. It provides a reference for better research of drugs to prevent and treat cerebral ischemia injury through the " nose-brain"pathway and lays a foundation for further research of the " nose-brain" pathway.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Medicina Tradicional Chinesa , Preparações Farmacêuticas , Administração Intranasal , Encéfalo , Sistemas de Liberação de Medicamentos , Humanos , Mucosa Nasal
5.
PLoS One ; 9(2): e89464, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586797

RESUMO

Epigenetic gene silencing by histone modifications and DNA methylation is essential for cancer development. The molecular mechanism that promotes selective epigenetic changes during tumorigenesis is not understood. We report here that the PIAS1 SUMO ligase is involved in the progression of breast tumorigenesis. Elevated PIAS1 expression was observed in breast tumor samples. PIAS1 knockdown in breast cancer cells reduced the subpopulation of tumor-initiating cells, and inhibited breast tumor growth in vivo. PIAS1 acts by delineating histone modifications and DNA methylation to silence the expression of a subset of clinically relevant genes, including breast cancer DNA methylation signature genes such as cyclin D2 and estrogen receptor, and breast tumor suppressor WNT5A. Our studies identify a novel epigenetic mechanism that regulates breast tumorigenesis through selective gene silencing.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Mama/patologia , Carcinogênese/genética , Epigênese Genética/genética , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Animais , Linhagem Celular Tumoral , Ciclina D2/genética , Metilação de DNA/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Humanos , Camundongos SCID , Proteínas Proto-Oncogênicas/genética , Receptores de Estrogênio/genética , Ubiquitina-Proteína Ligases/genética , Proteínas Wnt/genética , Proteína Wnt-5a
6.
EMBO J ; 33(2): 101-13, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24357619

RESUMO

The selective and temporal DNA methylation plays an important role in the self-renewal and differentiation of hematopoietic stem cells (HSCs), but the molecular mechanism that controls the dynamics of DNA methylation is not understood. Here, we report that the PIAS1 epigenetic pathway plays an important role in regulating HSC self-renewal and differentiation. PIAS1 is required for maintaining the quiescence of dormant HSCs and the long-term repopulating capacity of HSC. Pias1 disruption caused the abnormal expression of lineage-associated genes. Bisulfite sequencing analysis revealed the premature promoter demethylation of Gata1, a key myeloerythroid transcription factor and a PIAS1-target gene, in Pias1(-/-) HSCs. As a result, Pias1 disruption caused the inappropriate induction of Gata1 in HSCs and common lymphoid progenitors (CLPs). The expression of other myeloerythroid genes was also enhanced in CLPs and lineage-negative progenitors, with a concurrent repression of B cell-specific genes. Consistently, Pias1 disruption caused enhanced myeloerythroid, but reduced B lymphoid lineage differentiation. These results identify a novel role of PIAS1 in maintaining the quiescence of dormant HSCs and in the epigenetic repression of the myeloerythroid program.


Assuntos
Diferenciação Celular/genética , Proliferação de Células , Células-Tronco Hematopoéticas/fisiologia , Proteínas Inibidoras de STAT Ativados/fisiologia , Animais , Células da Medula Óssea/fisiologia , Linhagem da Célula/genética , Movimento Celular/genética , Microambiente Celular/genética , Epigênese Genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nicho de Células-Tronco/genética
8.
Science ; 330(6003): 521-5, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20966256

RESUMO

CD4(+)Foxp3(+) regulatory T (T(reg)) cells are important for maintaining immune tolerance. Understanding the molecular mechanism that regulates T(reg) differentiation will facilitate the development of effective therapeutic strategies against autoimmune diseases. We report here that the SUMO E3 ligase PIAS1 restricts the differentiation of natural T(reg) cells by maintaining a repressive chromatin state of the Foxp3 promoter. PIAS1 acts by binding to the Foxp3 promoter to recruit DNA methyltransferases and heterochromatin protein 1 for epigenetic modifications. Pias1 deletion caused promoter demethylation, reduced histone H3 methylation at Lys(9), and enhanced promoter accessibility. Consistently, Pias1(-/-) mice displayed an increased natural T(reg) cell population and were resistant to the development of experimental autoimmune encephalomyelitis. Our studies have identified an epigenetic mechanism that negatively regulates the differentiation of natural T(reg) cells.


Assuntos
Epigênese Genética , Linfopoese/genética , Proteínas Inibidoras de STAT Ativados/fisiologia , Proteínas Repressoras/fisiologia , Linfócitos T Reguladores/citologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Sítios de Ligação , Linfócitos T CD4-Positivos/citologia , Cromatina/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Encefalomielite Autoimune Experimental/imunologia , Feminino , Fatores de Transcrição Forkhead/genética , Histonas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Regiões Promotoras Genéticas , Linfócitos T Reguladores/imunologia , DNA Metiltransferase 3B
10.
Trends Pharmacol Sci ; 29(10): 505-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18755518

RESUMO

Protein sumoylation is a post-translational-modification event, in which small ubiquitin-like modifier (SUMO) is covalently attached to protein substrates by a three-step process. Sumoylation has been suggested to regulate multiple cellular processes, including inflammation. Inflammation is initiated in response to pathogenic infections, but uncontrolled inflammatory responses can lead to the development of inflammatory disorders such as rheumatoid arthritis. Recent studies indicate that proinflammatory stimuli, such as tumor necrosis factor alpha and lipopolysaccharide, can activate PIAS1 [protein inhibitor of activated STAT1 (signal transducer and activator of transcription 1)] SUMO E3 ligase through a SUMO-dependent, inhibitor of kappaB kinase alpha (IKKalpha)-mediated phosphorylation event. Activated PIAS1 is then recruited to inflammatory gene promoters to repress transcription. These findings support a hypothesis that therapies targeting the PIAS1 SUMO ligase pathway might be developed for the treatment of inflammatory disorders such as rheumatoid arthritis and atherosclerosis.


Assuntos
Inflamação/tratamento farmacológico , Proteínas Inibidoras de STAT Ativados/efeitos dos fármacos , Proteína SUMO-1/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Sistemas de Liberação de Medicamentos , Humanos , Inflamação/fisiopatologia , NF-kappa B/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Fator de Transcrição STAT1/metabolismo , Proteína SUMO-1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
11.
Curr Opin Cell Biol ; 20(3): 288-93, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18468876

RESUMO

Protein sumoylation has emerged as an important regulatory mechanism for the transcriptional machinery. Sumoylation is a highly dynamic process that is regulated in response to cellular stimuli or pathogenic challenges. Altered activity of the small ubiquitin-like modifier (SUMO) conjugation system is associated with human cancers and inflammation. Thus, understanding the regulation of protein sumoylation is important for the design of SUMO-based therapeutic strategies for the treatment of human diseases. Recent studies indicate that the sumoylation system can be regulated through multiple mechanisms, including the regulation of the expression of various components of the sumoylation pathway, and the modulation of the activity of SUMO enzymes. In addition, extracellular stimuli can signal the nucleus to trigger the rapid promoter recruitment of SUMO E3 ligases, resulting in the immediate repression of transcription. Finally, the sumoylation system can also be regulated through crosstalk with other post-translational modifications, including phosphorylation, ubiquitination, and acetylation.


Assuntos
Regulação da Expressão Gênica/genética , Proteínas Repressoras/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Transcrição Gênica/genética , Animais , Humanos , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Processamento de Proteína Pós-Traducional/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
12.
Cell ; 133(1): 103-15, 2008 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-18394993

RESUMO

RanBP2 is a nucleoporin with SUMO E3 ligase activity that functions in both nucleocytoplasmic transport and mitosis. However, the biological relevance of RanBP2 and the in vivo targets of its E3 ligase activity are unknown. Here we show that animals with low amounts of RanBP2 develop severe aneuploidy in the absence of overt transport defects. The main chromosome segregation defect in cells from these mice is anaphase-bridge formation. Topoisomerase IIalpha (Topo IIalpha), which decatenates sister centromeres prior to anaphase onset to prevent bridges, fails to accumulate at inner centromeres when RanBP2 levels are low. We find that RanBP2 sumoylates Topo IIalpha in mitosis and that this modification is required for its proper localization to inner centromeres. Furthermore, mice with low amounts of RanBP2 are highly sensitive to tumor formation. Together, these data identify RanBP2 as a chromosomal instability gene that regulates Topo IIalpha by sumoylation and suppresses tumorigenesis.


Assuntos
Antígenos de Neoplasias/metabolismo , Centrômero/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Chaperonas Moleculares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Anáfase , Aneuploidia , Animais , Carcinógenos , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Mitose , Chaperonas Moleculares/genética , Mutação , Neoplasias/induzido quimicamente , Neoplasias/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Estrutura Terciária de Proteína , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
13.
J Neurosci ; 27(37): 10037-46, 2007 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-17855618

RESUMO

Postsynaptic morphogenesis of dendrites is essential for the establishment of neural connectivity in the brain, but the mechanisms that govern postsynaptic dendritic differentiation remain poorly understood. Sumoylation of the transcription factor myocyte enhancer factor 2A (MEF2A) promotes the differentiation of postsynaptic granule neuron dendritic claws in the cerebellar cortex. Here, we identify the protein PIASx as a MEF2 SUMO E3 ligase that represses MEF2-dependent transcription in neurons. Gain-of-function and genetic knockdown experiments in rat cerebellar slices and in the postnatal cerebellum in vivo reveal that PIASx drives the differentiation of granule neuron dendritic claws in the cerebellar cortex. MEF2A knockdown suppresses PIASx-induced dendritic claw differentiation, and expression of sumoylated MEF2A reverses PIASx knockdown-induced loss of dendritic claws. These findings define the PIASx-MEF2 sumoylation signaling link as a key mechanism that orchestrates postsynaptic dendritic claw morphogenesis in the cerebellar cortex and suggest novel functions for SUMO E3 ligases in brain development and plasticity.


Assuntos
Dendritos/enzimologia , Proteínas de Domínio MADS/fisiologia , Morfogênese/fisiologia , Fatores de Regulação Miogênica/fisiologia , Proteínas Inibidoras de STAT Ativados/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/fisiologia , Linhagem Celular , Células Cultivadas , Cerebelo/enzimologia , Cerebelo/crescimento & desenvolvimento , Dendritos/fisiologia , Humanos , Proteínas de Domínio MADS/deficiência , Proteínas de Domínio MADS/genética , Fatores de Transcrição MEF2 , Morfogênese/genética , Fatores de Regulação Miogênica/genética , Proteínas Inibidoras de STAT Ativados/deficiência , Proteínas Inibidoras de STAT Ativados/genética , Ratos , Ratos Long-Evans , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sinapses/enzimologia , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
14.
Proc Natl Acad Sci U S A ; 104(28): 11643-8, 2007 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-17606919

RESUMO

NF-kappaB and STATs regulate multiple cellular processes through the transcriptional activation of genes with diversified functions. Although the molecular mechanisms that can turn on/off the overall NF-kappaB/STAT signaling have been extensively studied, how NF-kappaB/STAT-target genes can be differentially regulated is poorly understood. Here we report that PIASy, a member of the PIAS (for protein inhibitor of activated STAT) protein family, is a physiologically important transcriptional repressor of NF-kappaB and STAT1. Piasy deletion in dendritic cells resulted in enhanced expression of a subset of NF-kappaB and STAT1-dependent genes in response to LPS or IFN-gamma treatment, respectively. Consistently, Piasy null mice are hypersensitive to the LPS-induced endotoxic shock. Furthermore, PIASy and PIAS1 display specific as well as redundant effects on the regulation of NF-kappaB/STAT1 signaling. Pias1-/-Piasy-/- embryos died before day 11.5. The disruption of one allele of Pias1 in the Piasy-/- background significantly enhanced the effect of Piasy deletion on the transcriptional induction of NF-kappaB/STAT1-dependent genes, and vice versa. Our results demonstrate that PIASy cooperates with PIAS1 to regulate the specificity and magnitude of NF-kappaB/STAT1-mediated gene activation.


Assuntos
Regulação para Baixo/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas Inibidoras de STAT Ativados/fisiologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Proteínas Inibidoras de STAT Ativados/deficiência , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Fator de Transcrição STAT1/antagonistas & inibidores , Ativação Transcricional
15.
Cell ; 129(5): 903-14, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17540171

RESUMO

How inflammatory stimuli signal to the nucleus to restrict inflammation is poorly understood. Protein inhibitor of activated STAT1 (PIAS1), a transcriptional regulator that possesses small ubiquitin-related modifier (SUMO) E3 ligase activity, inhibits immune responses by selectively blocking the binding of NF-kappaB and STAT1 to gene promoters. We report here that PIAS1 becomes rapidly phosphorylated on Ser90 residue in response to various inflammatory stimuli. Mutational studies indicate that Ser90 phosphorylation is required for PIAS1 to repress transcription. Upon TNF treatment, wild-type PIAS1, but not the Ser90A mutant, becomes rapidly associated with the promoters of NF-kappaB target genes. Furthermore, IKKalpha, but not IKKbeta, interacts with PIAS1 in vivo and mediates PIAS1 Ser90 phosphorylation, a process that requires the SUMO ligase activity of PIAS1. Our results identify a signaling pathway in which proinflammatory stimuli activate the IKKalpha-mediated sumoylation-dependent phosphorylation of PIAS1 for the immediate repression of inflammatory gene activation.


Assuntos
Quinase I-kappa B/metabolismo , Inflamação/imunologia , Proteínas Inibidoras de STAT Ativados/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Expressão Gênica , Humanos , Quinase I-kappa B/genética , Ligantes , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Baço/citologia , Baço/imunologia , Baço/metabolismo , Transcrição Gênica
16.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 38(1): 64-7, 2007 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-17294730

RESUMO

OBJECTIVE: To investigate the expression of minichromosome maintenance protein 6 (MCM6) in tissue sections of craniopharyngioma and observe its relation with the outcome of patients with craniopharyngiomas. METHODS: Prospective cohorts were composed of 32 adamantine epithelioma (AE) patients and 31 squamous papillary tumor (SP) patients. The average of follow-up phase was 84. 26 months, of 60 patients with craniopharyngioma, 20 suffered from recurrence and underwent operation again for removal of tumor, and the specimens of the tumors patients were collected. MCM6 as proliferative marker expression in the specimen sections was measured by immunohistochemical method (avidin-biotin-peroxidase); quantitatively, scoring for MCM6 protein variation was performed by TE2000-U inverted biological microscope and Image-Pro Plus professional image analysis software. Oncocyte proliferation potential was evaluated for inter-group comparison in three pair of groups, including AE/SP, recurrence/recurrence-free, and primary/relapse groups. RESULTS: 14 of 32 AE patients and 6 of 31 SP patients had recurrence during follow-up. MCM6 protein expression showed significant difference between AE/SP groups and between recurrence/recurrence-free groups (P < 0.05, two-tailed), but there was no statistically significant difference between primary and recurrent craniopharyngiomas. CONCLUSION: The subtype and MCM6 protein expression in craniopharyngiomas are related to the prognosis of tumor and thus may be useful in predicting the risk of tumor relapse.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Craniofaringioma/diagnóstico , Craniofaringioma/genética , Regulação Neoplásica da Expressão Gênica , Animais , Craniofaringioma/patologia , Seguimentos , Humanos , Componente 6 do Complexo de Manutenção de Minicromossomo , Prognóstico , Recidiva
17.
Nat Cell Biol ; 9(1): 80-5, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17159996

RESUMO

Protein-tyrosine phosphatase 1B (PTP1B) is an ubiquitously expressed enzyme that negatively regulates growth-factor signalling and cell proliferation by binding to and dephosphorylating key receptor tyrosine kinases, such as the insulin receptor. It is unclear how the activity of PTP1B is regulated. Using a yeast two-hybrid assay, a protein inhibitor of activated STAT1 (PIAS1) was isolated as a PTP1B-interacting protein. Here, we show that PIAS1, which functions as a small ubiquitin-like modifier (SUMO) E3 ligase, associates with PTP1B in mammalian fibroblasts and catalyses sumoylation of PTP1B. Sumoylation of PTP1B reduces its catalytic activity and inhibits the negative effect of PTP1B on insulin receptor signalling and on transformation by the oncogene v-crk. Insulin-stimulated sumoylation of endogenous PTP1B results in a transient downregulation of the enzyme; this event does not occur when the endogenous enzyme is replaced with a sumoylation-resistant mutant of PTP1B. These results suggest that sumoylation, which has been implicated primarily in processes in the nucleus and nuclear pore, also modulates a key enzyme-substrate signalling complex that regulates metabolism and cell proliferation.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Tirosina Fosfatases/genética , Proteína SUMO-1/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Camundongos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Proteínas Inibidoras de STAT Ativados/isolamento & purificação , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/metabolismo , Transdução de Sinais , Transfecção
18.
EMBO J ; 25(11): 2358-67, 2006 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-16710296

RESUMO

Interferons (IFNs) regulate diverse cellular functions through activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway. Lack of Ubp43, an IFN-inducible ISG15 deconjugating enzyme, leads to IFN hypersensitivity in ubp43-/- mice, suggesting an important function of Ubp43 in downregulation of IFN responses. Here, we show that Ubp43 negatively regulates IFN signaling independent of its isopeptidase activity towards ISG15. Ubp43 functions specifically for type I IFN signaling by downregulating the JAK-STAT pathway at the level of the IFN receptor. Using molecular, biochemical, and genetic approaches, we demonstrate that Ubp43 specifically binds to the IFNAR2 receptor subunit and inhibits the activity of receptor-associated JAK1 by blocking the interaction between JAK and the IFN receptor. These data implicate Ubp43 as a novel in vivo inhibitor of signal transduction pathways that are specifically triggered by type I IFN.


Assuntos
Citocinas/metabolismo , Endopeptidases/metabolismo , Interferon Tipo I/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Citocinas/genética , Regulação para Baixo , Endopeptidases/genética , Humanos , Janus Quinase 1 , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Tirosina Quinases/metabolismo , Receptor de Interferon alfa e beta , Receptores de Interferon/metabolismo , Fatores de Transcrição STAT/metabolismo , Ubiquitina Tiolesterase , Ubiquitinas/genética , Ubiquitinas/metabolismo
19.
Cell Res ; 16(2): 196-202, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16474434

RESUMO

Cytokines activate multiple signal transduction pathways to regulate gene expression. STATs and NF-kB are two important families of transcription factors activated by cytokines. Abnormal regulation of STAT and NF-kB activities has been associated with human diseases. The protein inhibitor of activated STAT (PIAS) protein family has been proposed to interact with over 60 proteins, many of which are transcription factors involved in the immune system. PIAS proteins regulate transcription through several mechanisms, including blocking the DNA-binding activity of transcription factors, recruiting transcriptional co-repressors and promoting protein sumoylation. This article is to review the role of PIAS proteins in the regulation of STAT and NF-kB signaling pathways.


Assuntos
Citocinas/metabolismo , Regulação da Expressão Gênica , Proteínas Inibidoras de STAT Ativados/metabolismo , Transdução de Sinais/fisiologia , Citocinas/genética , Humanos , Janus Quinases/metabolismo , NF-kappa B/metabolismo , Proteínas Inibidoras de STAT Ativados/genética , Fatores de Transcrição STAT/metabolismo
20.
Nat Rev Immunol ; 5(8): 593-605, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16056253

RESUMO

The protein inhibitor of activated STAT (PIAS) family of proteins has been proposed to regulate the activity of many transcription factors, including signal transducer and activator of transcription proteins (STATs), nuclear factor-kappaB, SMA- and MAD-related proteins (SMADs), and the tumour-suppressor protein p53. PIAS proteins regulate transcription through several mechanisms, including blocking the DNA-binding activity of transcription factors, recruiting transcriptional corepressors or co-activators, and promoting protein sumoylation. Recent genetic studies support an in vivo function for PIAS proteins in the regulation of innate immune responses. In this article, we review the current understanding of the molecular basis, specificity and physiological roles of PIAS proteins in the regulation of gene-activation pathways in the immune system.


Assuntos
Citocinas/metabolismo , Regulação da Expressão Gênica , Sistema Imunitário/metabolismo , Proteínas/metabolismo , Animais , Apoptose/genética , Ciclo Celular/genética , Citocinas/genética , Humanos , Proteínas Inibidoras de STAT Ativados , Estrutura Secundária de Proteína , Proteínas/química , Transdução de Sinais/genética , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA