Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Biochem Biophys Res Commun ; 467(4): 778-84, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26482848

RESUMO

The candidate tumor suppressor gene RASSF1A (Ras-association domain family 1, isoform A) is inactivated in many types of adult and childhood cancers. However, the mechanisms by which RASSF1A exerts tumor suppressive functions have yet to be elucidated. In this report, we sought to identify candidate proteins that interact with RASSF1A using proteomic screening. Using peptide mass fingerprinting, we identified protein arginine N-methyltransferase 5 (PRMT5), a type II protein arginine N-methyltransferase that monomethylates and symmetrically dimethylates arginine residues, as a novel protein that interacts with RASSF1A. The association between the two proteins was confirmed by co-immunoprecipitation and immunofluorescence staining. Co-expressing RASSF1A and PRMT5 led to a redistribution of PRMT5 from the cytosol to stabilized microtubules, where RASSF1A and PRMT5 became co-localized. Our results demonstrate that PRMT5 translocates to bundled microtubules on stabilization by RASSF1A expression. Our results show that the tumor suppressor RASSF1A interacts with PRMT5 in vivo and in vitro. Notably, this is the first demonstration of RASSF1A-dependent microtubule recruitment of PRMT5, suggesting a novel role for RASSF1A in the anchoring of cytosolic PRMT5 to microtubules.


Assuntos
Microtúbulos/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Células COS , Chlorocebus aethiops , Citosol/metabolismo , Células HEK293/metabolismo , Humanos , Transporte Proteico , Proteína-Arginina N-Metiltransferases/genética , Proteômica/métodos , Proteínas Supressoras de Tumor/genética
2.
Ann Surg Oncol ; 19(2): 612-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21913013

RESUMO

BACKGROUND: Patients with clinical stage I/II (T1-2N0M0) oral tongue squamous cell carcinoma (TSCC) usually undergo partial glossectomy alone. However, 14-48% of them develop delayed neck metastasis (DNM), which may lead to an unfavorable course. Recently epithelial-to-mesenchymal transition (EMT) has been thought to play a crucial role in cancer metastasis. The present study aimed to examine the associations of EMT-involved molecular factors and clinicopathological factors with DNM in stage I/II TSCC. METHODS: mRNA expression levels of E-cadherin and its transcriptional repressors (snail, SIP1, and twist) in 7 head and neck squamous cell carcinoma (HNSCC) cell lines were evaluated by quantitative real-time PCR. Clinicopathological parameters and immunohistochemical expressions of E-cadherin and its repressors were examined in surgical specimens of 37 stage I/II TSCC patients who underwent partial glossectomy alone. RESULTS: In HNSCC cells, E-cadherin expression was inversely correlated with SIP1 expression (P = 0.023). Univariate analysis of immunohistochemistry showed that overexpression of SIP1 and loss of E-cadherin were significantly correlated with DNM, although no inverse correlation was found between E-cadherin and its repressors. Multiple logistic regression analysis including clinicopathological and molecular factors revealed that overexpression of SIP1 (P = 0.005), loss of E-cadherin (P = 0.046), and vascular invasion (P = 0.024) were independently correlated with DNM. CONCLUSIONS: These results suggest that development of DNM in stage I/II TSCC is closely related to induction of EMT in primary tumor cells. Especially, SIP1 and E-cadherin are considered to be the possible markers for selecting patients at high risk of DNM.


Assuntos
Biomarcadores Tumorais/genética , Caderinas/genética , Glossectomia , Excisão de Linfonodo , Proteínas do Tecido Nervoso/genética , Proteínas de Ligação a RNA/genética , Neoplasias da Língua/genética , Neoplasias da Língua/cirurgia , Biomarcadores Tumorais/metabolismo , Western Blotting , Caderinas/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/cirurgia , Regulação para Baixo , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Masculino , Prontuários Médicos , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Proteínas do Tecido Nervoso/metabolismo , Prognóstico , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias da Língua/metabolismo , Células Tumorais Cultivadas
3.
Curr Biol ; 31(8): 1581-1591.e3, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33651990

RESUMO

The spindle-assembly checkpoint facilitates mitotic fidelity by delaying anaphase onset in response to microtubule vacancy at kinetochores. Following microtubule attachment, kinetochores receive microtubule-derived force, which causes kinetochores to undergo repetitive cycles of deformation; this phenomenon is referred to as kinetochore stretching. The nature of the forces and the relevance relating this deformation are not well understood. Here, we show that kinetochore stretching occurs within a framework of single end-on attached kinetochores, irrespective of microtubule poleward pulling force. An experimental method to conditionally interfere with the stretching allowed us to determine that kinetochore stretching comprises an essential process of checkpoint silencing by promoting PP1 phosphatase recruitment after the establishment of end-on attachments and removal of the majority of checkpoint-activating kinase Mps1 from kinetochores. Remarkably, we found that a lower frequency of kinetochore stretching largely correlates with a prolonged metaphase in cancer cell lines with chromosomal instability. Perturbation of kinetochore stretching and checkpoint silencing in chromosomally stable cells produced anaphase bridges, which can be alleviated by reducing chromosome-loaded cohesin. These observations indicate that kinetochore stretching-mediated checkpoint silencing provides an unanticipated etiology underlying chromosomal instability and underscores the importance of a rapid metaphase-to-anaphase transition in sustaining mitotic fidelity.


Assuntos
Segregação de Cromossomos , Cinetocoros , Pontos de Checagem da Fase M do Ciclo Celular , Fuso Acromático , Anáfase , Linhagem Celular Tumoral , Instabilidade Cromossômica , Humanos , Microtúbulos
4.
Biochem Biophys Res Commun ; 392(4): 603-7, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20100459

RESUMO

Cellular adaptations to chronic opioid treatment result in enhanced responsiveness of adenylate cyclase and an increase in forskolin- or agonist-stimulated cAMP production. It is, however, not known whether chaperone molecules such as heat shock proteins contribute to this adenylate cyclase sensitization. Here, we report that treatment of cells with geldanamycin, an inhibitor of heat shock protein 90 (Hsp90), led to effective attenuation of morphine-induced adenylate cyclase sensitization. In SK-N-SH human neuroblastoma cells, morphine significantly increased RNA transcript and protein levels of type I adenylate cyclase, leading to sensitization. Whole-genome tiling array analysis revealed that cAMP response element-binding protein, an important mediator for cellular adaptation to morphine, associated with the proximal promoter of Hsp90AB1 not only in SK-N-SH cells but also in rat PC12 and human embryonic kidney cells. Hsp90AB1 transcript and protein levels increased significantly during morphine treatment, and co-application of geldanamycin (0.1-10 nM) effectively suppressed the increase in forskolin-activated adenylate cyclase activation by 56%. Type I adenylate cyclase, but not Hsp90AB1, underwent significant degradation during geldanamycin treatment. These results indicate that Hsp90 is a new pharmacological target for the suppression of adenylate cyclase sensitization induced by chronic morphine treatment.


Assuntos
Adenilil Ciclases/biossíntese , Tolerância a Medicamentos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Morfina/farmacologia , Entorpecentes/farmacologia , Adenilil Ciclases/genética , Animais , Benzoquinonas/farmacologia , Linhagem Celular Tumoral , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Lactamas Macrocíclicas/farmacologia , Ratos , Receptores Opioides mu/genética
5.
Am J Cancer Res ; 8(10): 2046-2063, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30416855

RESUMO

Tumor cell-derived vascular endothelial growth factor (VEGF)-C has been primarily implicated in promoting lymphangiogenesis by activating Flt-4 (VEGFR-3) expressed on lymphatic endothelial cells via a paracrine mechanism. Flt4 has also been shown to be expressed selectively in subsets of cancer cells. However, little is known about the functional role of VEGF-C/Flt4 signaling via an autocrine mechanism, as well as the clinicopathological implication of the VEGF-C/Flt4 axis and its downstream effector molecules, in head and neck squamous cell carcinoma (HNSCC), including oral squamous cell carcinoma (OSCC). In the present study, we detected Flt-4 expression selectively in several HNSCC cell lines by quantitative PCR, and its internalization reflecting receptor activation was confirmed by immunocytochemistry in SAS and HO1U1 cells. Flt-4 stimulation upregulated the expression of contactin-1 (CNTN-1, a neural cell adhesion molecule) and VEGF-C itself in SAS cells, while Flt-4 inhibition downregulated the expression of CNTN-1 in both SAS and HO1U1 cells and that of VEGF-C itself in SAS cells. In vitro cell proliferation and migration assays using SAS cells demonstrated that both cell proliferation and migration were promoted by Flt-4 stimulation, while those were suppressed by Flt-4 inhibition. Clinicopathological factors and immunohistochemical expression of Flt-4, VEGF-C, and CNTN-1 in tumor cells were evaluated using surgical specimens from patients with tongue squamous cell carcinoma. We found a significant correlation of CNTN-1 expression with both VEGF-C and Flt-4 expression, but not between VEGF-C and Flt-4. Multivariate logistic regression analysis revealed that T classification (P = 0.003), lymphatic invasion (P = 0.024), and Flt-4 expression in tumor cells (P = 0.046) were independently predictive of neck lymph node metastasis. These results suggest that the VEGF-C/Flt-4 axis in tumor cells enhances tumor cell proliferation and migration via upregulating the expression of VEGF-C itself and CNTN-1 in an autocrine manner, thereby contributing to cancer progression of OSCC, including neck metastasis. Hence, targeting the VEGF-C/Flt-4 axis in tumor cells can be an attractive therapeutic strategy for the treatment of cancer.

6.
Nat Neurosci ; 21(6): 820-833, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29713080

RESUMO

Chronic morphine exposure upregulates adenylate cyclase signaling and reduces analgesic efficacy, a condition known as opioid tolerance. Nonopioid neurotransmitters can enhance morphine tolerance, but the mechanism for this is poorly understood. We show that morphine tolerance was delayed in mice lacking vasopressin 1b receptors (V1bRs) or after administration of V1bR antagonist into the rostral ventromedial medulla, where transcripts for V1bRs and µ-opioid receptors are co-localized. Vasopressin increased morphine-binding affinity in cells expressing both V1bR and µ-opioid receptors. Complex formation among V1bR, ß-arrestin-2, and µ-opioid receptor resulted in vasopressin-mediated upregulation of ERK phosphorylation and adenylate cyclase sensitization. A leucine-rich segment in the V1bR C-terminus was necessary for the association with ß-arrestin-2. Deletion of this leucine-rich segment increased morphine analgesia and reduced vasopressin-mediated adenylate cyclase sensitization. These findings indicate that inhibition of µ-opioid-receptor-associated V1bR provides an approach for enhancing morphine analgesia without increasing analgesic tolerance.


Assuntos
Tolerância a Medicamentos/genética , Morfina/farmacologia , Entorpecentes/farmacologia , Receptores Opioides mu/metabolismo , Receptores de Vasopressinas/metabolismo , beta-Arrestina 2/metabolismo , Adenilil Ciclases/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Injeções , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Bulbo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/farmacocinética , Dependência de Morfina/psicologia , Entorpecentes/farmacocinética , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Fosforilação , Receptores Opioides mu/genética , Receptores de Vasopressinas/genética , beta-Arrestina 2/genética
7.
J Inflamm (Lond) ; 13: 7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26949374

RESUMO

BACKGROUND: The amount of urinary glycoprotein lipocalin 2 (LCN2) has been known to increase after kidney injury because of failed reabsorption by the proximal tubules or direct secretion from injured tissues. However, the relationship between urinary tract obstruction and the isoform diversity of LCN2 has not been examined. METHODS: The urinary levels of LCN2 isoforms were examined in male mice after an intraperitoneal injection of lipopolysaccharide (LPS) or in a mouse model of unilateral ureter obstruction (UUO). The LCN2 levels in sera, bladder urine, renal pelvic urine, and tissue samples were also analyzed. Endo- and exoglycosidases were used to investigate the different N-glycan patterns of LCN2. RESULTS: Two isoforms of urinary LCN2 with different molecular weights were identified in an immunoblotting analysis, and the levels of both isoforms were increased 6 h after LPS administration. The primary LCN2 isoform was the lower molecular weight 22-kDa isoform, which was detected in the serum, urine, liver and kidney. In contrast, the 24-kDa LCN2 isoform was detected only in urine. In the UUO experiments, the levels of the 24-kDa LCN2 were increased in the bladder urine but not in the urine accumulated in the renal pelvis due to UUO. The 22-kDa LCN2 was identified in the renal pelvic urine from UUO mice. The peptide-N glycosidase F digestion of the two urinary LCN2 isoforms generated a single protein. Moreover, the two urinary LCN2 proteins were sensitive to neuraminidase and resistant to endoglycosidase H (Endo H). The LCN2 in the serum, lung and kidney was resistant to Endo H, as observed in urine, whereas the LCN2 in the liver and the ureter were degraded by this enzyme. CONCLUSIONS: These results suggest that the difference in the molecular weights of the LCN2 proteins was due to their N-glycan structure. The high molecular weight LCN2 in urine could be detected after the inflammatory response to LPS and UUO. Furthermore, the sensitivity to Endo H identified the presence of two types of carbohydrate moieties, depending on the tissue in which the LCN2 was produced. These findings are useful for widening the clinical applicability of urinary LCN2 analyses.

8.
Trends Endocrinol Metab ; 14(3): 107-13, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12670735

RESUMO

Alpha(1)-adrenoceptors (ARs) mediate some of the main actions of the natural catecholamines, epinephrine and norepinephrine, and have a crucial role in the regulation of arterial blood pressure. Since alpha(1)-AR was subdivided into three subtypes (alpha(1A)-AR, alpha(1B)-AR and alpha(1D)-AR), the search has been on to discover subtype-specific physiological roles and to develop subtype-selective agonists and antagonists. Recently, several strains of genetically engineered mice have become available. Studies with these mice have provided several clues to help elucidate subtype-specific physiological functions; for instance, alpha(1A)-AR and alpha(1D)-AR subtypes play an important role in the regulation of blood pressure, suggesting that subtype-selective antagonists might be desirable antihypertensive agents. The ability to study subtype-specific functions in different mouse strains by altering the same alpha(1)-AR in different ways strengthens the conclusions drawn from pharmacological studies. Although these genetic approaches have limitations, they have significantly increased our understanding of the functions of alpha(1)-AR subtypes.


Assuntos
Receptores Adrenérgicos alfa 1/fisiologia , Animais , Doenças Cardiovasculares/fisiopatologia , Fenômenos Fisiológicos Cardiovasculares , Sistema Nervoso Central/fisiologia , Doenças do Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptores Adrenérgicos alfa 1/classificação , Receptores Adrenérgicos alfa 1/genética , Resistência Vascular/genética
9.
Eur J Pharmacol ; 765: 291-9, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26318147

RESUMO

Only limited information is available on agonist-dependent changes in the subcellular localization of vasopressin V1B receptors. Our radioligand binding study of membrane preparations and intact cells revealed that a large fraction of the V1B receptor is located in the cytoplasm in unstimulated CHO cells, which is in contrast to the plasma membrane localization of the V1A and V2 receptors. Moreover, when the affinity of radiolabeled arginine-vasopressin ([3H]AVP) was compared between membrane preparations and intact cells, the affinity of [3H]AVP to the cell surface V1B receptors, but not the V1A receptors, was significantly reduced. Although the number and affinity of cell surface V1B receptors decreased, they became extensively internalized upon binding with [3H]AVP. Approximately 87% of cell surface-bound [3H]AVP was internalized and became resistant to acid wash during incubation with 1 nM [3H]AVP. By contrast, less ligand (35%) was internalized in the cells expressing the V1A receptor. Extensive internalization of the V1B receptors was partially attenuated by inhibitors of cytoskeletal proteins, siRNA against ß-arrestin 2, or the removal of sodium chloride from the extracellular buffer, indicating that this internalization involves clathrin-coated pits. Together, these results indicate that the mechanism that regulates the number and affinity of V1B receptors in the plasma membrane is markedly distinct from the corresponding mechanisms for the V1A and V2 receptors and plays a critical role under stress conditions, when vasopressin release is augmented.


Assuntos
Arginina Vasopressina/farmacologia , Membrana Celular/metabolismo , Espaço Intracelular/metabolismo , Hipófise/metabolismo , Receptores de Vasopressinas/agonistas , Animais , Sítios de Ligação , Células CHO , Técnicas de Cultura de Células , Cricetulus , Células HEK293 , Humanos , Camundongos , Transporte Proteico , Ensaio Radioligante , Ratos Endogâmicos WKY , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Transfecção , Trítio
10.
Eur J Pharmacol ; 749: 98-106, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25592317

RESUMO

A group of synthetic substance P (SP) antagonists, such as [Arg(6),D-Trp(7,9),N(Me)Phe(8)]-substance P(6-11) and [D-Arg(1),D-Phe(5),D-Trp(7,9),Leu(11)]-substance P, bind to a range of distinct G-protein-coupled receptor (GPCR) family members, including V1a vasopressin receptors, and they competitively inhibit agonist binding. This extended accessibility enabled us to identify a GPCR subset with a partially conserved binding site structure. By combining pharmacological data and amino acid sequence homology matrices, a pharmacological lineage of GPCRs that are sensitive to these two SP antagonists was constructed. We found that sensitivity to the SP antagonists was not limited to the Gq-protein-coupled V1a and V1b receptors; Gs-coupled V2 receptors and oxytocin receptors, which couple with both Gq and Gi, also demonstrated sensitivity. Unexpectedly, a dendrogram based on the amino acid sequences of 222 known GPCRs showed that a group of receptors sensitive to the SP antagonists are located in close proximity to vasopressin/oxytocin receptors. Gonadotropin-releasing peptide receptors, located near the vasopressin receptors in the dendrogram, were also sensitive to the SP analogs, whereas α1B adrenergic receptors, located more distantly from the vasopressin receptors, were not sensitive. Our finding suggests that pharmacological lineage analysis is useful in selecting subsets of candidate receptors that contain a conserved binding site for a ligand with broad-spectrum binding abilities. The knowledge that the binding site of the two broad-spectrum SP analogs partially overlaps with that of distinct peptide agonists is valuable for understanding the specificity/broadness of peptide ligands.


Assuntos
Receptores de Ocitocina/metabolismo , Receptores de Vasopressinas/metabolismo , Substância P/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Hormônio Liberador de Gonadotropina/metabolismo , Peptídeos/metabolismo , Receptores de Ocitocina/química , Receptores de Vasopressinas/química , Homologia de Sequência , Substância P/análogos & derivados
11.
J Exp Clin Cancer Res ; 33: 40, 2014 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-24887090

RESUMO

BACKGROUND: The epithelial-to-mesenchymal transition (EMT) accompanied by the downregulation of E-cadherin has been thought to promote metastasis. Cyclooxygenase-2 (Cox-2) is presumed to contribute to cancer progression through its multifaceted function, and recently its inverse relationship with E-cadherin was suggested. The aim of the present study was to investigate whether selective Cox-2 inhibitors restore the expression of E-cadherin in head and neck squamous cell carcinoma (HNSCC) cells, and to examine the possible correlations of the expression levels of EMT-related molecules with clinicopathological factors in HNSCC. METHODS: We used quantitative real-time PCR to examine the effects of three selective Cox-2 inhibitors, i.e., celecoxib, NS-398, and SC-791 on the gene expressions of E-cadherin (CDH-1) and its transcriptional repressors (SIP1, Snail, Twist) in the human HNSCC cell lines HSC-2 and HSC-4. To evaluate the changes in E-cadherin expression on the cell surface, we used a flowcytometer and immunofluorescent staining in addition to Western blotting. We evaluated and statistically analyzed the clinicopathological factors and mRNA expressions of Cox-2, CDH-1 and its repressors in surgical specimens of 40 patients with tongue squamous cell carcinoma (TSCC). RESULTS: The selective Cox-2 inhibitors upregulated the E-cadherin expression on the cell surface of the HNSCC cells through the downregulation of its transcriptional repressors. The extent of this effect depended on the baseline expression levels of both E-cadherin and Cox-2 in each cell line. A univariate analysis showed that higher Cox-2 mRNA expression (p = 0.037), lower CDH-1 mRNA expression (p = 0.020), and advanced T-classification (p = 0.036) were significantly correlated with lymph node metastasis in TSCC. A multivariate logistic regression revealed that lower CDH-1 mRNA expression was the independent risk factor affecting lymph node metastasis (p = 0.041). CONCLUSIONS: These findings suggest that the appropriately selective administration of certain Cox-2 inhibitors may have an anti-metastatic effect through suppression of the EMT by restoring E-cadherin expression. In addition, the downregulation of CDH-1 resulting from the EMT may be closely involved in lymph node metastasis in TSCC.


Assuntos
Caderinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Transição Epitelial-Mesenquimal , Neoplasias Bucais/metabolismo , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Antígenos CD , Antineoplásicos/farmacologia , Caderinas/genética , Carcinoma de Células Escamosas/secundário , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/patologia , Análise Multivariada , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Língua/metabolismo
12.
Eur J Pharmacol ; 670(1): 208-15, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21933671

RESUMO

Effective detection of potential nephrotoxicity is crucial for pre-clinical drug development. We evaluated a sensitive animal model for drug-induced kidney injury, which includes hemi-nephrectomy of mice. Although bucillamine and d-penicillamine are used for the treatment of rheumatoid arthritis in Japan, drug-related adverse effects on the kidney can limit their therapeutic utilities. When bucillamine (1000 or 2000 mg/kg/day) or d-penicillamine (2000 mg/kg/day) were orally administered to hemi-nephrectomised BALB/c mice, the urinary protein levels of bucillamine-treated mice, but not of those treated with d-penicillamine, the vehicle, or in bucillamine treated unnephrectomized mice, were significantly increased and remained high during the 4-week drug-loading period. Membranous glomerulonephropathy occasionally seen in bucillamine/d-penicillamine-treated arthritis patients was not reproduced in mice. Instead, our mouse model showed proximal tubular injury and podocyte foot process effacement in the bucillamine-treated kidneys. These two cell types are also the primary targets of the experimental Heymann membranous glomerulonephropathy. Gene expression profiling of the bucillamine-treated mice identified lipocalin 2 as a significantly up-regulated transcript together with cytochrome P450 CYP4a14, a group-specific component, and proprotein convertase subtilisin/kexin type 9. Moreover, large amounts of lipocalin 2 were detected in the urine of the bucillamine-treated mice, but not in the hemi-nephrectomised control mice. These results indicate that hemi-nephrectomy effectively promotes acute kidney injury by bucillamine, which is accompanied by up-regulation of the urinary biomarker lipocalin 2. Our mouse model with initial stage of kidney injury should be useful to analyse the pathogenesis of drug-induced glomerular and tubular injuries.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Cisteína/análogos & derivados , Túbulos Renais Proximais/efeitos dos fármacos , Podócitos/efeitos dos fármacos , Injúria Renal Aguda/sangue , Injúria Renal Aguda/genética , Injúria Renal Aguda/urina , Animais , Cisteína/toxicidade , Modelos Animais de Doenças , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nefrectomia , Análise de Sequência com Séries de Oligonucleotídeos , Podócitos/metabolismo , Podócitos/patologia , Proteinúria/sangue , Proteinúria/induzido quimicamente , Proteinúria/genética , Proteinúria/urina , Transcriptoma/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
13.
Life Sci ; 86(11-12): 455-60, 2010 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-20149803

RESUMO

AIMS: A noncoding RNA, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), is upregulated in several malignant tumors. Its expression in neuroblastoma, however, is not known, and the regulatory mechanisms of MALAT1 gene expression have not been elucidated. The aim of this study is to clarify how MALAT1 gene expression is altered by extracellular signals in the SK-N-SH neuroblastoma cell line and to define its proximal promoter in order to study the mechanism of MALAT1 gene expression. METHODS: Transcript amounts were analyzed by real-time semiquantitative polymerase chain reaction (qPCR). Genes coregulated with MALAT1 were identified by DNA microarray analysis. The structure of the MALAT1 transcript was delineated using a tiling microarray, and the 5'-end was determined using the rapid amplification of cDNA ends (RACE) method. We investigated binding of the cyclic AMP-responsive element binding (CREB) transcription factor to the MALAT1 promoter by using chromatin immunoprecipitation (ChIP) followed by tiling array analysis, and the results were confirmed using ChIP-qPCR. KEY FINDINGS: The posterior pituitary hormone oxytocin increased the levels of MALAT1 and immediate early gene transcripts as early as 15 min after stimulation. Although the expression of immediate early genes returned to basal levels after 3h, MALAT1 transcript levels peaked 6-24h after stimulation. We identified a shorter transcriptional initiation site and found that CREB binds to the defined proximal promoter of the MALAT1 gene. SIGNIFICANCE: The expression of the tumor marker MALAT1 ncRNA is sensitive to cell surface receptor activation by oxytocin in a neuroblastoma cell line.


Assuntos
Biomarcadores Tumorais/biossíntese , Ocitocina/farmacologia , RNA Neoplásico/biossíntese , Biomarcadores Tumorais/análise , Neoplasias Encefálicas/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/biossíntese , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Humanos , Informática , Neuroblastoma/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Neoplásico/análise , Receptores de Ocitocina/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Biol Chem ; 279(39): 40807-18, 2004 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-15272025

RESUMO

Insulin-like growth factor-I (IGF-I) plays a role in mutually exclusive processes such as proliferation and differentiation in a variety of cell types. IGF-I is a potent mitogen and motogen for dedifferentiated vascular smooth muscle cells (VSMCs) in vivo and in vitro. However, in differentiated VSMCs, IGF-I is only required for maintaining the differentiated phenotype. Here we investigated the VSMC phenotype-dependent signaling and biological processes triggered by IGF-I. In differentiated VSMCs, IGF-I activated a protein-tyrosine phosphatase, SHP-2, recruited by insulin receptor substrate-1 (IRS-1). The activated SHP-2 then dephosphorylated IRS-1 Tyr(P)-895, resulting in blockade of the pathways from IRS-1/Grb2/Sos to the ERK and p38 MAPK. Conversely, such negative regulation was silent in dedifferentiated VSMCs, where IGF-I activated both MAPKs via IRS-1/Grb2/Sos interaction-linked Ras activation, leading to proliferation and migration. Thus, our present results demonstrate that the IRS-1/SHP-2 interaction acts as a switch controlling VSMC phenotype-dependent IGF-I-induced signaling pathways and biological processes, and this mechanism is likely to be applicable to other cells.


Assuntos
Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Fosfoproteínas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , Divisão Celular , Movimento Celular , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Fenótipo , Fosforilação , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo , Tirosina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno , Proteínas ras/metabolismo
15.
J Biol Chem ; 278(1): 672-8, 2003 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-12409310

RESUMO

Three distinct subtypes of alpha(1)-adrenergic receptors (alpha(1)A-, alpha(1)B-, and alpha(1)D-AR) play a prominent role in cell growth. However, little is known about subtype-specific effects on cell proliferation. The activation of alpha(1)A- or alpha(1)B-AR inhibits serum-promoted cell proliferation, whereas alpha(1)D-AR activation does not show such an inhibitory effect. Notably, cell-cycle progression was blocked at G(1)/S transition after activation of alpha(1)A/alpha(1)B-AR but not of alpha(1)D-AR. In agreement with the differential cell proliferation effect, cAMP production was increased after activation of alpha(1)A/alpha(1)B-AR but not alpha(1)D-AR, whereas all alpha(1)-AR subtypes are associated with inositol 1,4,5-trisphosphate production and mitogen-activated protein kinase activation in a similar fashion. Furthermore, the serum-induced reduction in the levels of the cyclin-dependent kinase inhibitor, p27(Kip1), was blocked after activation of alpha(1)A/alpha(1)B-AR but not alpha(1)D-AR. These results show that alpha(1)-AR subtypes differentially activate the cAMP/p27(Kip1) pathway and thereby have differential inhibitory effects on cell proliferation. Subtype-dependent effects should be taken into consideration when assessing the physiological response of native cells where alpha(1)-AR subtypes are generally co-expressed.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , AMP Cíclico/metabolismo , Isoformas de Proteínas/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Células CHO , Cricetinae , Meios de Cultura , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Citometria de Fluxo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Isoformas de Proteínas/genética , Receptores Adrenérgicos alfa 1/genética , Transdução de Sinais/fisiologia
16.
Biochem Biophys Res Commun ; 299(1): 142-7, 2002 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-12435400

RESUMO

We have established that differing effects of alpha1-adrenergic receptor (AR) subtypes on cell proliferation are due to differential coupling to the Gs/cAMP pathway; thus, both alpha1A- and alpha1B-ARs couple to Gs, while alpha1D-AR does not. To identify the region responsible for this difference in subtype-specific Gs coupling, we constructed a series of chimeric and a set of point-mutated human alpha1A- and alpha1D-ARs, and examined their signaling ability. Here, we show that the amino acid residues Thr 136 and Val138 in the intracellular loop II of the human alpha1A-AR are intimately involved with Gs coupling.


Assuntos
Aminoácidos/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Sequência de Aminoácidos , Divisão Celular , Membrana Celular , AMP Cíclico/metabolismo , Humanos , Ligantes , Dados de Sequência Molecular , Mutação , Ligação Proteica , Ensaio Radioligante , Receptores Adrenérgicos alfa 1/química , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Treonina/metabolismo , Transfecção , Valina/metabolismo
17.
Am J Physiol Heart Circ Physiol ; 285(1): H194-203, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12793978

RESUMO

Mutations in cardiac voltage-gated K+ channels cause long QT syndrome (LQTS) and sudden death. We created a transgenic mouse with a long QT phenotype (Kv1DN) by overexpression of a truncated K+ channel in the heart and investigated whether the dominant negative effect of the transgene would be overcome by the direct injection of adenoviral vectors expressing wild-type Kv1.5 (AV-Kv1.5) into the myocardium. End points at 3-10 days included electrophysiology in isolated cardiomyocytes, surface ECG, programmed stimulation of the right ventricle, and in vivo optical mapping of action potentials and repolarization gradients in Langendorff-perfused hearts. Overexpression of Kv1.5 reconstituted a 4-aminopyridine-sensitive outward K+ current, shortened the action potential duration, eliminated early afterdepolarizations, shortened the QT interval, decreased dispersion of repolarization, and increased the heart rate. Each of these changes is consistent with a physiologically significant primary effect of adenoviral expression of Kv1.5 on ventricular repolarization of Kv1DN mice.


Assuntos
Síndrome do QT Longo/fisiopatologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , 4-Aminopiridina/farmacologia , Potenciais de Ação/fisiologia , Adenoviridae/genética , Animais , Western Blotting , Células Cultivadas , Clonagem Molecular , Eletrocardiografia , Eletrofisiologia , Técnicas de Transferência de Genes , Canal de Potássio Kv1.5 , Síndrome do QT Longo/genética , Camundongos , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa