Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 259: 115043, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37224781

RESUMEN

Cigarette smoking is an independent risk factor for lung cancer. Nicotine, as an addictive substance in tobacco and e-cigarettes, is known to promote tumor progression and metastasis despite being a non-carcinogen. As a tumor suppressor gene, JWA is widely involved in the inhibition of tumor growth and metastasis and the maintenance of cellular homeostasis, including in non-small cell lung cancer (NSCLC). However, the role of JWA in nicotine-induced tumor progression remains unclear. Here, we reported for the first time that JWA was significantly downregulated in smoking-related lung cancer and associated with overall survival. Nicotine exposure reduced JWA expression in a dose-dependent manner. Gene Set Enrichment Analysis (GSEA) analysis showed the tumor stemness pathway was enriched in smoking-related lung cancer, and JWA was negatively associated with stemness molecules CD44, SOX2, and CD133. JWA also inhibited nicotine-enhanced colony formation, spheroid formation, and EDU incorporation in lung cancer cells. Mechanically, nicotine downregulated JWA expression via the CHRNA5-mediated AKT pathway. Lower JWA expression enhanced CD44 expression through inhibition of ubiquitination-mediated degradation of Specificity Protein 1 (SP1). The in vivo data indicated that JAC4 through the JWA/SP1/CD44 axis inhibited nicotine-triggered lung cancer progression and stemness. In conclusion, JWA via down-regulating CD44 inhibited nicotine-triggered lung cancer cell stemness and progression. Our study may provide new insights to develop JAC4 for the therapy of nicotine-related cancers.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Sistemas Electrónicos de Liberación de Nicotina , Neoplasias Pulmonares , Receptores Nicotínicos , Humanos , Neoplasias Pulmonares/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Nicotina/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Receptores de Hialuranos/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores Nicotínicos/metabolismo , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo
2.
Int J Mol Sci ; 24(10)2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37240137

RESUMEN

Lung adenocarcinoma (LUAD) is the most common lung cancer, with high mortality. As a tumor-suppressor gene, JWA plays an important role in blocking pan-tumor progression. JAC4, a small molecular-compound agonist, transcriptionally activates JWA expression both in vivo and in vitro. However, the direct target and the anticancer mechanism of JAC4 in LUAD have not been elucidated. Public transcriptome and proteome data sets were used to analyze the relationship between JWA expression and patient survival in LUAD. The anticancer activities of JAC4 were determined through in vitro and in vivo assays. The molecular mechanism of JAC4 was assessed by Western blot, quantitative real-time PCR (qRT-PCR), immunofluorescence (IF), ubiquitination assay, co-immunoprecipitation, and mass spectrometry (MS). Cellular thermal shift and molecule-docking assays were used for confirmation of the interactions between JAC4/CTBP1 and AMPK/NEDD4L. JWA was downregulated in LUAD tissues. Higher expression of JWA was associated with a better prognosis of LUAD. JAC4 inhibited LUAD cell proliferation and migration in both in-vitro and in-vivo models. Mechanistically, JAC4 increased the stability of NEDD4L through AMPK-mediated phosphorylation at Thr367. The WW domain of NEDD4L, an E3 ubiquitin ligase, interacted with EGFR, thus promoting ubiquitination at K716 and the subsequent degradation of EGFR. Importantly, the combination of JAC4 and AZD9191 synergistically inhibited the growth and metastasis of EGFR-mutant lung cancer in both subcutaneous and orthotopic NSCLC xenografts. Furthermore, direct binding of JAC4 to CTBP1 blocked nuclear translocation of CTBP1 and then removed its transcriptional suppression on the JWA gene. The small-molecule JWA agonist JAC4 plays a therapeutic role in EGFR-driven LUAD growth and metastasis through the CTBP1-mediated JWA/AMPK/NEDD4L/EGFR axis.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Línea Celular Tumoral , Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proliferación Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica
3.
Biochem Biophys Res Commun ; 521(3): 753-761, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31703842

RESUMEN

Two Pt(IV) prodrugs, Cx-platin-Cl and Cx-DN604-Cl, derived from the conjugation of cisplatin or DN604 with a CK2 inhibitor CX-4945, were constructed to suppress DNA damage repair-related elements. During in vitro biological studies, the Pt(IV) prodrugs had excellent cytotoxicity superior to cisplatin and DN604 to reverse drug resistance. Further mechanistic investigations revealed that the powerful anticancer activity of Cx-platin-Cl and Cx-DN604-Cl arisen from its suppression of JWA-XRCC1-mediated single-strand breaks repair. The emerging Pt(IV) prodrugs inhibited the growth of the xenografted tumors of C57BL6 and nude mice apart from JWA-/- mice. Between them, Cx-platin-Cl augmented the infiltration and proliferation of Teff cells, alleviated the recruitment of Treg cells. The results provided compelling preclinical support that Cx-platin-Cl and Cx-DN604-Cl could reverse chemo-immune resistance via decaying JWA-XRCC1-mediated SSBR and immunosuppression, improving the development of emerging Pt(IV) candidate as a potential immunotherapeutic agent for cancer resistant prevention.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas de Choque Térmico/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Naftiridinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Compuestos Organoplatinos/uso terapéutico , Profármacos/uso terapéutico , Animales , Antineoplásicos/química , Línea Celular Tumoral , Roturas del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Desnudos , Naftiridinas/química , Neoplasias/inmunología , Neoplasias/metabolismo , Compuestos Organoplatinos/química , Fenazinas , Profármacos/química , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1552-1561, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29481911

RESUMEN

Gastric cancer (GC) is one of the most common malignant cancers worldwide. Metastasis leads to poor prognoses in GC patients in advanced stages. Our previous studies have demonstrated that JWA functions as a tumour suppressor and that low expression of JWA in GC tissues is significantly correlated with shorter overall survival (OS) as well as with advanced clinicopathologic features in patients. However, the mechanism of dysregulation of JWA in cancers is not clear. In the present study, we found that an E3 ubiquitin ligase, RNF185, directly interacted with JWA and promoted its ubiquitination at the K158 site, resulting in subsequent degradation. Moreover, the protein level of RNF185 was negatively correlated with JWA in tumour tissues from GC patients. High RNF185 expression was significantly correlated with shorter OS. Additionally, increased RNF185 expression facilitated GC cell migration in vitro and promoted GC metastasis in vivo by downregulating JWA expression. However, this effect was reversed by replenishment of JWA. In conclusion, our findings highlight the following: (1) RNF185 promotes GC metastasis by mediating JWA degradation via a ubiquitin-proteasome pathway; (2) the K158 site of JWA is essential for its ubiquitination in GC cells. These findings suggest that RNF185 is a novel candidate prognostic marker and potential therapeutic target for GC.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Gástricas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Línea Celular Tumoral , Movimiento Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas de Choque Térmico/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Mitocondriales/genética , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Ubiquitina-Proteína Ligasas/genética
5.
Zhonghua Yi Xue Za Zhi ; 97(27): 2141-2144, 2017 Jul 18.
Artículo en Zh | MEDLINE | ID: mdl-28763891

RESUMEN

Objective: To explore the effect of JWA on cisplatin sensitivity and its potential molecular mechanism in esophageal cancer. Methods: The siRNA was used to inhibit the JWA expression, then cisplatin sensitivity and LC3 (autophagy related protein) expression levels were observed in TE1 cells.Further, the effect of autophagy inhibitor tamoxifen (3-MA) on above process was determined.Cisplatin sensitivity of 20 fresh esophageal cancer samples was evaluated by histoculture drug response assay (HDRA). Result: Silencing JWA gene increased the sensitivity of TE1 cells to cisplatin (P<0.05), and decreased the LC3-Ⅰ and LC3-Ⅱ proteins induced by cisplatin.Furthermore, combined with 3-MA increased the inhibition rate of cisplatin in JWA silencing group (P<0.05). Additionally, the inhibition rate of cisplatin on tissues with low JWA expression were higher than those with high expression (45.6% vs 25.6%, P=0.005). Conclusions: JWA could influence the cisplatin sensitivity by regulating autophagy in esophageal cancer.


Asunto(s)
Autofagia , Apoptosis , Línea Celular Tumoral , Cisplatino , Neoplasias Esofágicas , Humanos
6.
Mol Carcinog ; 53(4): 325-36, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23169062

RESUMEN

We previously identified JWA as a novel microtubule-associated protein (MAP), which is implicated in carcinogenesis and tumor progression. The aims of the present study were to investigate the biological action and the prognostic significance of JWA in hepatocellular carcinoma (HCC). Quantitative real-time PCR and Western blot were used to detect JWA mRNA and protein expression, respectively, in stepwise metastatic HCC cell lines and HCC tissues. Short hairpin RNA was used to inhibit JWA expression in HCC cells. The effects of JWA depletion on cell migration, invasion, adhesion and in vivo metastasis were investigated. Immunohistochemistry of JWA was conducted in microarrays with tissue from 314 HCC patients who had undergone surgical resection. Prognostic significance was assessed using the Kaplan-Meier method and log-rank tests. The result showed JWA expression was decreased in the highly metastatic HCC cell lines and HCC tissues. Depletion of JWA caused a notable increase in cell migration, invasion and adhesion in vitro and metastasis in vivo. Furthermore, there was an inverse correlation between JWA expression and FAK expression and phosphorylation, RhoA activation and matrix metalloproteinase-2 (MMP-2) activity in HCC cells. More notably, multivariate analysis revealed that a low level of JWA expression was an independent prognosticator for both recurrence-free and overall survival for HCC patients after surgical resection, especially for AFP-normal HCC patients. Taken together, our data demonstrate that JWA plays a crucial role in HCC progression and suggest JWA may be a potential prognostic biomarker and therapeutic target for HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Regulación hacia Abajo , Proteínas de Choque Térmico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Secuencia de Bases , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/enzimología , Línea Celular Tumoral , Cartilla de ADN , Activación Enzimática , Proteína-Tirosina Quinasas de Adhesión Focal/química , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Neoplasias Hepáticas/enzimología , Metaloproteinasas de la Matriz/metabolismo , Proteínas de Transporte de Membrana , Fosforilación , Reacción en Cadena de la Polimerasa , Pronóstico
7.
Redox Biol ; 73: 103190, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38744191

RESUMEN

Parkinson's disease (PD) poses a significant challenge in neurodegenerative disorders, characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). The intricate mechanisms orchestrating DA neurodegeneration in PD are not fully understood, necessitating the exploration of innovative therapeutic approaches. Recent studies have implicated ferroptosis as a major contributor to the loss of DA neurons, revealing a complex interplay between iron accumulation and neurodegeneration. However, the sophisticated nature of this process challenges the conventional belief that mere iron removal could effectively prevent DA neuronal ferroptosis. Here, we report JWA, alternatively referred to as ARL6IP5, as a negative regulator of ferroptosis, capable of ameliorating DA neuronal loss in the context of PD. In this study, synchronized expression patterns of JWA and tyrosine hydroxylase (TH) in PD patients and mice were observed, underscoring the importance of JWA for DA neuronal survival. Screening of ferroptosis-related genes unraveled the engagement of iron metabolism in the JWA-dependent inhibition of DA neuronal ferroptosis. Genetic manipulation of JWA provided compelling evidence linking its neuroprotective effects to the attenuation of NCOA4-mediated ferritinophagy. Molecular docking, co-immunoprecipitation, and immunofluorescence studies confirmed that JWA mitigated DA neuronal ferroptosis by occupying the ferritin binding site of NCOA4. Moreover, the JWA-activating compound, JAC4, demonstrated promising neuroprotective effects in cellular and animal PD models by elevating JWA expression, offering a potential avenue for neuroprotection in PD. Collectively, our work establishes JWA as a novel regulator of ferritinophagy, presenting a promising therapeutic target for addressing DA neuronal ferroptosis in PD.


Asunto(s)
Neuronas Dopaminérgicas , Ferritinas , Ferroptosis , Coactivadores de Receptor Nuclear , Enfermedad de Parkinson , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/genética , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Animales , Ratones , Humanos , Coactivadores de Receptor Nuclear/metabolismo , Coactivadores de Receptor Nuclear/genética , Ferritinas/metabolismo , Ferritinas/genética , Hierro/metabolismo , Modelos Animales de Enfermedad , Unión Proteica , Autofagia , Masculino
8.
Cancers (Basel) ; 14(19)2022 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-36230577

RESUMEN

Tumor heterogeneity limits the precision treatment of targeted drugs. It is important to find new tumor targets. JWA, also known as ADP ribosylation factor-like GTPase 6 interacting protein 5 (ARL6IP5, GenBank: AF070523, 1998), is a microtubule-associated protein and an environmental response gene. Substantial evidence shows that JWA is low expressed in a variety of malignancies and is correlated with overall survival. As a tumor suppressor, JWA inhibits tumor progression by suppressing multiple oncogenes or activating tumor suppressor genes. Low levels of JWA expression in tumors have been reported to be associated with multiple aspects of cancer progression, including angiogenesis, proliferation, apoptosis, metastasis, and chemotherapy resistance. In this review, we will discuss the structure and biological functions of JWA in tumors, examine the potential therapeutic strategies for targeting JWA and explore the directions for future investigation.

9.
Int J Biol Sci ; 18(14): 5503-5521, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36147468

RESUMEN

The intestinal epithelium is a rapid self-renewal and regenerated tissue of which the structural integrity is beneficial for maintaining health. The integrity of intestinal epithelium depends on the balance of cell proliferation, differentiation, migration, and the function of intestinal stem cells, which declines due to genetic defect or aging. Jwa participates in multiple cellular processes; it also responds to oxidative stress and repairs DNA damage. However, whether Jwa plays a role in maintaining the homeostasis of intestinal renewal and regeneration is not clear. In the present study, we firstly described that the deletion of Jwa disturbed the homeostasis of intestinal epithelial renewal and regeneration. Jwa deficiency promoted NOTCH1 degradation in the ERK/FBXW7-mediated ubiquitin-proteasome pathway, thus disturbing the PPARγ/STAT5 axis. These mechanisms might partially contribute to the reduction of intestinal stem cell function and alteration of intestinal epithelial cell lineage distribution, finally suppressing the renewal and regeneration of intestinal epithelium. Moreover, our results also revealed that Jwa was a novel putative aging related gene.


Asunto(s)
PPAR gamma , Factor de Transcripción STAT5 , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Proteínas de Choque Térmico/genética , Homeostasis/genética , Mucosa Intestinal/metabolismo , Proteínas de Transporte de Membrana/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factor de Transcripción STAT5/metabolismo , Ubiquitinas/metabolismo
10.
Int J Clin Exp Pathol ; 13(12): 3120-3127, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33425112

RESUMEN

Gastric cancer is one of the common gastrointestinal tumors. Tumor recurrence leads to a high death rate of gastric cancer. It is very important to find markers to effectively predict gastric cancer recurrence. We constructed a gastric cancer tissue microarray containing 89 tumors and corresponding normal tissues to explore the relationship between some proteins' expression and gastric cancer recurrence. The expression of JWA, Cullin1, p53, XRCC1, CHIP, FAK, MMP-2, MDM2 and p21 was determined on the microarray by immunohistochemistry. The relationship between the expression of these proteins and gastric cancer recurrence was analyzed. Tumor diameter, lymph node metastasis, and TNM stage were closely related with gastric cancer recurrence by Fisher's exact test (P<0.05). We used the univariate Cox regression analysis to find that JWA, XRCC1 were related to gastric cancer recurrence (P<0.05); Lymph node metastasis and TNM stage were closely related to gastric cancer recurrence (P<0.05). Multivariate Cox regression analysis revealed that XRCC1 or lymph node metastasis were independent risk factors of gastric cancer recurrence (P<0.05). Kaplan-Meier survival curve assay indicated that patients with low JWA or XRCC1 expression in gastric cancer had significantly shorter DFS than those with high-expressed proteins (P<0.05). JWA or XRCC1 may be effective markers to predict gastric cancer recurrence.

11.
Exp Ther Med ; 15(4): 3509-3515, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29545876

RESUMEN

The present study aimed to investigate the effects of JWA knockout (JWA-/-) on malignant transformation of murine embryonic fibroblast (MEF) cells using a conditional JWA-/- mouse model. Once MEF cells were prepared, the potential role of JWA-/- on proliferation, migration, invasion and colony formation of MEF cells was investigated by cytological examination. The effects of JWA-/- on the regulation and protein expression levels of epithelial-mesenchymal transition (EMT)-related proteins in MEF cells, including poly(ADP-ribose) polymerase-1 (PARP-1), vimentin, ß-catenin and E-cadherin, were investigated using western blot analysis. The tumorigenicity of JWA deficiency was explored using nude mouse xenografts and subcutaneous inoculation of MEF cells exhibiting JWA-/-. JWA-/- was able to increase cell proliferation, migration, invasion and colony formation in the malignant transformation of MEF cells. The protein expression levels of PARP-1, vimentin and ß-catenin were upregulated, whereas E-cadherin was downregulated in JWA-/- MEF cells. The tumor formation was observed in mice following subcutaneous inoculation of MEF with JWA-/-, whereas no tumor was formed in the mice treated with functional JWA MEF cells. In conclusion, the present findings suggest that JWA-/- has important roles in cell proliferation, migration, invasion and colony formation and is able to induce the malignant transformation of MEF cells. The expression levels of EMT-related proteins changed and tumorigenicity increased in JWA-/- MEF cells compared with cells with functional JWA. The present findings indicate that JWA may function as an anti-oncogene in tumorigenesis.

12.
Toxicol Lett ; 277: 32-40, 2017 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-28428137

RESUMEN

Paraquat (PQ), a widely used environmental toxin in agriculture, contributes to the onset and progression of Parkinson's disease (PD) by damaging neurons. The JWA gene, also known as ARL6IP5, exerts a protective effect on degenerating dopamine (DA) neurons. However, the roles of JWA in PQ-induced neuronal damage are still unknown. In our study, two neuronal cell lines (HT-22 and SH-SY5Y) and neuron-specific JWA knockout (JWA-nKO) and age-matched wild-type (JWA-nWT) mice were subjected to PQ treatment. The results indicate that PQ administration triggers the upregulation of JWA. Elevated expression of JWA rescues the accumulation of reactive oxygen species (ROS) while increasing glutathione (GSH) and glutathione peroxidase (GPx) levels under PQ exposure. Further investigations revealed that the protective effect of JWA mostly involves regulation of the MEK/PI3K-Nrf2 axis. Our results suggest that JWA may be a novel target for the prevention and treatment of PQ-induced PD.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Proteínas de Choque Térmico/metabolismo , Herbicidas/toxicidad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Paraquat/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Proteínas Portadoras/genética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Células HEK293 , Proteínas de Choque Térmico/genética , Humanos , Concentración 50 Inhibidora , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas de Transporte de Membrana , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Neuronas/metabolismo , Neuronas/patología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética , Activación Transcripcional , Transfección
13.
Biochem Biophys Rep ; 11: 72-78, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28955770

RESUMEN

Addicsin (Arl6ip5) is a multifunctional physiological and pathophysiological regulator that exerts its effects by readily forming homo- and hetero-complexes with various functional factors. In particular, addicsin acts as a negative modulator of neural glutamate transporter excitatory amino acid carrier 1 (EAAC1) and participates in the regulation of intracellular glutathione (GSH) content by negatively modulating EAAC1-mediated cysteine and glutamate uptake. Addicsin is considered to play a crucial role in the onset of neurodegenerative diseases including epilepsy. However, the molecular dynamics of addicsin remains largely unknown. Here, we report the dynamics of addicsin in NG108-15 cells upon exposure to pentylenetetrazol (PTZ), a representative epileptogenic agent acting on the gamma-Aminobutyric acid A (GABAA) receptor. Fluorescent immunostaining analysis demonstrated that addicsin drastically changed its localization from the endoplasmic reticulum (ER) to the plasma membrane within 1 h of PTZ exposure in a dose-dependent manner. Moreover, addicsin was co-localized with the plasma membrane markers EAAC1 and Na+/K+ ATPase alpha-3 upon PTZ stimulation. This translocation was significantly inhibited by a non-competitive GABAA receptor antagonist, picrotoxin, but not by a competitive GABAA receptor antagonist, bicuculline. Furthermore, lactate dehydrogenase (LDH) assay and 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical-scavenging assay showed that PTZ-induced addicsin translocation was accompanied by a decrease of radical-scavenging activity and an increase of cytotoxicity in a PTZ dose-dependent manner. These findings suggest that PTZ induces the translocation of addicsin from the ER to the plasma membrane and modulates the redox system by regulating EAAC1-mediated GSH synthesis, which leads to the activation of cell death signaling.

14.
Oncotarget ; 7(24): 36865-36884, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27167206

RESUMEN

BACKGROUND AND AIMS: JWA, a microtubule-associated protein (MAP) involved in apoptosis, has been identified as a suppressor of metastasis, and it affects cell migration in melanoma and its downregulation in tumor is an idependent negative prognostic factor in resectable gastric cancer. HER2 overexpression has been observed in gastric cancer (GC) cells and implicated in the metastatic phenotype. However, the biological role of JWA in migration and its clinical value in HER2-positive GC remain elusive. RESULTS: JWA suppresses EGF-induced cell migration and actin cytoskeletal rearrangement by abrogating HER2 expression and downstream PI3K/AKT signaling in HER2-overexpressing GC cell lines. The modulation of HER2 by JWA is dependent on ERK activation and consequent PEA3 upregulation and activation. Reduced JWA expression is associated with high HER2 expression and with poor survival in patients with AGC, whereas HER2 expression alone is not associated with survival. However, concomitant low JWA and high HER2 expression is associated with unfavorable outcomes. Additionally, when patients were stratified by JWA expression, those with higher HER2 expression in the low JWA expression subgroup exhibited worse survival. METHODS: The impact of JWA on the EGF-induced migration of HER2-positive GC cells was studied using transwell assays and G-LISA assays. Western blotting, real-time PCR, electrophoretic mobility shift assays and luciferase assays were utilized to investigate the mechanisms by which JWA affects HER2. The association of JWA with HER2 and its clinical value were further analyzed by IHC in 128 pairs of advanced gastric cancer (AGC) and adjacent normal tissue samples. CONCLUSIONS: This study characterizes a novel mechanism for regulating cell motility in HER2-overexpressing GC cells involving JWA-mediated MEK/ERK/PEA3 signaling activation and HER2 downregulation. Furthermore, JWA may be a useful prognostic indicator for advanced GC and may help stratify HER2-positive patient subgroups to better identify unfavorable outcomes.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas de Choque Térmico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Gástricas/patología , Adulto , Anciano , Biomarcadores de Tumor/análisis , Movimiento Celular/fisiología , Citoesqueleto/patología , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Proteínas de Transporte de Membrana , Persona de Mediana Edad , Pronóstico , Receptor ErbB-2/biosíntesis , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad
15.
Mol Neurobiol ; 53(1): 355-368, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25432888

RESUMEN

JWA (Arl6ip5), a homologous gene of glutamate-transporter-associated protein 3-18 (GTRAP3-18) and addicsin, is highly expressed in hippocampus. We generated systemic and neuronal JWA knockout (JWA-KO and JWA-nKO) mice to investigate the influence of JWA deficiency on spatial cognitive performance, process of neurogenesis, and induction of long-term potentiation (LTP) in hippocampal dentate gyrus (DG). In comparison with wild-type (WT) mice and JWA (loxP/loxP) (control of JWA-nKO) mice, 8-week-old JWA-KO mice and JWA-nKO mice showed spatial cognitive potentiation as assessed by Morris water maze test. In hippocampal DG of JWA-nKO mice, either survival and migration or neurite growth of newborn neurons were significantly enhanced without the changes in proliferation and differentiation of stem cells. In addition, the increase of LTP amplitude and the decline of LTP threshold were observed in DG, but not in CA1 region, of JWA-nKO mice compared to control mice. The levels of hippocampal FAK, Akt, and mTOR phosphorylation in JWA-nKO mice were higher than those in control mice. The PI3K or FAK inhibitor could abolish the enhanced neurogenesis and LTP induction in JWA-nKO mice, which was accompanied by disappearance of the spatial cognitive potentiation. The treatment of JWA-nKO mice with 3'-azido-3'-deoxythymidine (AZT), a telomerase inhibitor, suppressed not only the enhanced neurogenesis but also the enhanced LTP induction in DG, but it did not affect the LTP induction in CA1 region. The results suggest that the JWA deficiency through cascading FAK-PI3K-Akt-mTOR pathway increases the newborn neurons and enhances the LTP induction in hippocampal DG, which leads to the spatial cognitive potentiation.


Asunto(s)
Proteínas Portadoras/metabolismo , Cognición , Giro Dentado/fisiología , Potenciación a Largo Plazo , Neurogénesis , Animales , Animales Recién Nacidos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cognición/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Genotipo , Proteínas de Choque Térmico , Potenciación a Largo Plazo/efectos de los fármacos , Proteínas de Transporte de Membrana , Ratones Noqueados , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neurogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Zidovudina/farmacología
16.
Oncotarget ; 7(44): 71790-71801, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27708243

RESUMEN

Human epidermal growth factor receptor 2 (HER2) targeted therapy is currently considered as the standard treatment for HER2-positive advanced gastric cancer (GC). However, unsatisfactory results of recent phase III clinical trials involving lapatinib suggested biomarkers for selection of patients. The aim of this study was to identify JWA as a biomarker for lapatinib resistance in GC cells and elucidate the underlying mechanisms. Lapatinib was effective to the intrinsic cisplatin-resistant GC cells. JWA activation conferred lapatinib unresponsiveness, but reversed cisplatin resistance in GC cells. Whereas, deletion of JWA significantly restored lapatinib suppression on proliferation and lapatinib-induced apoptosis. JWA-induced down-regulation of HER2 and activation of ERK phosphorylation led to lapatinib resistance. Furthermore, c-Cbl represented a novel mechanism for HER2 degradation enhanced by JWA in GC cells. Taken together, JWA is a potential predictive marker for lapatinib resistance, targeting the patients that may benefit from lapatinib treatment in human GC.


Asunto(s)
Proteínas de Choque Térmico/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Proto-Oncogénicas c-cbl/fisiología , Quinazolinas/uso terapéutico , Receptor ErbB-2/análisis , Neoplasias Gástricas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/uso terapéutico , Regulación hacia Abajo , Resistencia a Antineoplásicos , Humanos , Lapatinib , Proteínas de Transporte de Membrana , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/química , Neoplasias Gástricas/patología
17.
Oncol Lett ; 8(4): 1859-1863, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25202426

RESUMEN

The present study aimed to investigate the role of JWA gene in the proliferation, apoptosis, invasion and migration of PANC-1 pancreatic cancer cells and the effect on the MAPK signaling pathway. Human PANC-1 pancreatic cancer cells were cultured in vitro, and small interfering RNA (siRNA) was designed for the JWA gene. The siRNA was transfected into PANC-1 cells. Subsequently, the cell proliferation was measured by MTT assay; cell apoptosis was detected by analyzing BAX and Bcl-2 protein expression; cell migration and invasion were measured using Transwell® chambers; and the protein expression of JWA and ERK1/2, JNK and p38 and their phosphorylated forms were measured by western blotting. By utilizing the MTT assay, the results showed that when JWA protein expression was inhibited, the proliferation of PANC-1 cells was enhanced. In addition, the expression of apoptosis-associated protein (AAP) BAX was substantially decreased, while the expression of the apoptosis inhibitor gene, Bcl-2, was significantly enhanced. Using Transwell chambers, it was found that the number of penetrating PANC-1 cells was significantly increased after transfection with JWA siRNA, suggesting that the migration and invasion of the cells was substantially increased. By studying the association between JWA and the MAPK pathway in PANC-1 cells, it was found that the expression of p-ERK1/2 of the MAPK pathway was significantly downregulated following JWA siRNA transfection. However, the expression levels of ERK1/2, JNK, p38, p-JNK and p-p38 showed no significant differences. In conclusion, it was shown that JWA affects the proliferation, apoptosis, invasion and migration of PANC-1 pancreatic cancer cells which could be attributed to effects on the expression of ERK1/2 in the MAPK pathway.

18.
CNS Neurosci Ther ; 20(8): 754-62, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24628733

RESUMEN

AIMS: To investigate the role of astrocytic JWA expression in dopaminergic (DA) neuron degeneration and in the pathogenesis of Parkinson's disease (PD). METHODS: Conditional astrocytic JWA null (JWA∆2/∆2/GFAP-Cre) mice and U251 glioma cells were used to evaluate the effects of JWA gene on DA neuron degeneration. The oxidative stress-driven molecular events were determined in both in vivo and in vitro models. RESULTS: Conditional astrocytic JWA knockout resulted in significant activation of astrocytes measured by increase in glial fibrillary acidic protein-positive cells (1.34×10(3)±74.5 vs. 8.44×10(3)±1.35×10(3), P<0.01) in mouse substantia nigra, accompanied by loss of DA neurons (1.03×10(4)±238 vs. 6.17×10(3)±392, P<0.001). Deficiency of JWA significantly aggravated reactive oxygen species (ROS) accumulation in substantia nigra compared with the wild-type mice. Increasing JWA expression in U251 glioma cells inhibited ROS with a concomitant increase in intracellular glutathione. Furthermore, suppression of IKKß-nuclear factor (NF)-κB signaling pathway was shown to regulate JWA in a PD model. CONCLUSIONS: The JWA gene exerts neuroprotective roles against DA neuronal degeneration via modulating intracellular redox status and NF-κB signaling pathway and is a potential treatment target for PD.


Asunto(s)
Astrocitos/metabolismo , Proteínas Portadoras/metabolismo , Dopamina/metabolismo , Intoxicación por MPTP/patología , Neuronas/patología , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutatión/metabolismo , Proteínas de Choque Térmico , Ácido Homovanílico/metabolismo , Quinasa I-kappa B/metabolismo , Proteínas de Transporte de Membrana , Ratones , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sustancia Negra/patología , Transfección
19.
Exp Ther Med ; 7(6): 1767-1771, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24926382

RESUMEN

The aim of the present study was to investigate whether the JWA gene regulates the proliferation, migration and invasion of human esophageal squamous cell carcinoma (ESCC) and normal human esophageal cell lines through mitogen-activated protein kinase (MAPK) signal transduction pathways. The role of JWA in proliferation, migration, invasion and apoptosis was investigated in the Eca109 human ESCC and HET-1A normal human esophageal cell lines via transfection with JWA-small interfering (si)RNA. Western blot analysis was conducted to observe the effect of JWA on apoptosis and the regulatory effect of JWA on proliferation was determined using a thiazolyl blue tetrazolium bromide (MTT) assay. Cellular migration and invasion were analyzed via a Transwell assay. In addition, the expression levels of extracellular signal-regulated protein kinases 1 and 2 (ERK1/2), c-Jun N-terminal kinase (JNK) and p38 MAPK following JWA-siRNA transfection were detected by western blot analysis and compared with those of untreated cells. The downregulation of JWA protein decreased apoptosis and increased the proliferation, migration and invasion of the Eca109 and HET-1A cell lines. In the Eca109 cell line, the expression levels of phosphorylated (p)-ERK1/2 and p-JNK, but not those of p-p38, decreased significantly in the JWA siRNA group compared with those in the control groups. However, in the HET-1A cell line, JWA-siRNA transfection significantly inhibited the expression of p-p38 and demonstrated no effect on the expression levels of p-ERK1/2 and p-JNK. In conclusion, the JWA gene may regulate the ESCC and human esophageal cell lines through MAPK signaling pathways via different regulatory mechanisms.

20.
Neurochem Int ; 71: 22-35, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24680664

RESUMEN

Addicsin is a novel factor encoding a 23-kDa hydrophobic protein that is highly upregulated in the amygdala nuclei of morphine-administered mice. It is a murine homolog of human JWA and rat glutamate transporter-associated protein 3-18 (GTRAP3-18), a negative modulator of the neural glutamate transporter excitatory amino acid carrier 1 (EAAC1). Recent findings demonstrated that addicsin participates in various physiological processes by forming hetero- or homomultimeric complexes. However, the binding targets and molecular functions of addicsin remain largely unknown. To identify potential factors that associate with mouse addicsin, we performed a yeast two-hybrid screen using a 17-day-old mouse whole embryo cDNA library. We identified tomoregulin-1 (TR1) as a novel addicsin-associated factor. TR1, a type I transmembrane protein containing two follistatin-like modules and an epidermal growth factor-like domain, participates in nodal and bone morphogenetic protein signaling. Immunoprecipitation assays demonstrated that TR1 bound to addicsin, and that amino acids 145-188 of addicsin and amino acids 228-266 of TR1 were important for the formation of the addicsin-TR1 heterocomplex. The double-fluorescent immunohistochemical analysis revealed that addicsin and TR1 were coexpressed in neurons in the mature mouse brain regions tested. Moreover, TR1 showed a punctuate pattern throughout the cell, with preferential expression on the cell surface when expressed alone. However, TR1 predominantly redistributed to the endoplasmic reticulum (ER) when coexpressed with addicsin. Furthermore, coexpression of an addicsin mutant that lacked TR1 binding ability had little effect on the distribution of TR1. Biotinylation assays showed that coexpression of addicsin with TR1 suppressed the cell surface expression of TR1. Wound-healing assays demonstrated that the interaction of addicsin with TR1 had a significant effect on cell migration. These findings demonstrate that addicsin in the ER controls intracellular TR1 trafficking from the ER to plasma membrane and regulates cell migration through its interaction with TR1.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Animales , Biotinilación , Química Encefálica/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico , Masculino , Proteínas de Transporte de Membrana , Ratones , Plásmidos/genética , Receptores de Superficie Celular/metabolismo , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA