Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 131(2): 347-360, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38822145

RESUMEN

BACKGROUND: Sunitinib has emerged as the primary treatment for advanced or metastatic clear cell renal cell carcinoma (ccRCC) due to its significant improvement in patients' average survival time. However, drug resistance and adverse effects of sunitinib pose challenges to its clinical benefits. METHODS: The differentially expressed genes (DEGs) associated with sunitinib sensitivity and resistance in ccRCC were investigated. Cell counting kit-8, plate colony formation, flow cytometry and subcutaneous xenograft tumor model assays were employed to explore the effects of PDZK1 on ccRCC. Further research on the molecular mechanism was conducted through western blot, co-immunoprecipitation, immunofluorescence co-localization and immunohistochemical staining. RESULTS: We elucidated that PDZK1 is significantly downregulated in sunitinib-resistant ccRCC specimens, and PDZK1 negatively regulates the phosphorylation of PDGFR-ß and the activation of its downstream pathways through interaction with PDGFR-ß. The dysregulated low levels of PDZK1 contribute to inadequate inhibition of cell proliferation, tumor growth, and insensitivity to sunitinib treatment. Notably, our preclinical investigations showed that miR-15b antagomirs enhance sunitinib cytotoxic effects against ccRCC cells by upregulating PDZK1 levels, suggesting their potential in overcoming sunitinib resistance. CONCLUSIONS: Our findings establish the miR-15b/PDZK1/PDGFR-ß axis as a promising therapeutic target and a novel predictor for ccRCC patients' response to sunitinib treatment.


Asunto(s)
Carcinoma de Células Renales , Resistencia a Antineoplásicos , Neoplasias Renales , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Sunitinib , Sunitinib/farmacología , Sunitinib/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/metabolismo , Humanos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Animales , Resistencia a Antineoplásicos/genética , Ratones , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , MicroARNs/genética , Transducción de Señal/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino , Ratones Desnudos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
2.
Mol Cancer ; 21(1): 151, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35864508

RESUMEN

BACKGROUND: Recurrence and chemoresistance constitute the leading cause of death in colorectal cancer (CRC). Thus, it is of great significance to clarify the underlying mechanisms and identify predictors for tailoring adjuvant chemotherapy to improve the outcome of CRC. METHODS: By screening differentially expressed genes (DEGs), constructing random forest classification and ranking the importance of DEGs, we identified membrane associated guanylate kinase, WW and PDZ domain containing 3 (MAGI3) as an important gene in CRC recurrence. Immunohistochemical and western blot assays were employed to further detect MAGI3 expression in CRC tissues and cell lines. Cell counting kit-8, plate colony formation, flow cytometry, sub-cutaneous injection and azoxymethane plus dextran sulfate sodium induced mice CRC assays were employed to explore the effects of MAGI3 on proliferation, growth, cell cycle, apoptosis, xenograft formation and chemotherapy resistance of CRC. The underlying molecular mechanisms were further investigated through gene set enrichment analysis, quantitative real-time PCR, western blot, co-immunoprecipitation, ubiquitination, GST fusion protein pull-down and immunohistochemical staining assays. RESULTS: Our results showed that dysregulated low level of MAGI3 was correlated with recurrence and poor prognosis of CRC. MAGI3 was identified as a novel substrate-binding subunit of SKP1-Cullin E3 ligase to recognize c-Myc, and process c-Myc ubiquitination and degradation. Expression of MAGI3 in CRC cells inhibited cell growth, promoted apoptosis and chemosensitivity to fluoropyrimidine-based chemotherapy by suppressing activation of c-Myc in vitro and in vivo. In clinic, the stage II/III CRC patients with MAGI3-high had a significantly good recurrence-free survival (~ 80%, 5-year), and were not necessary for further adjuvant chemotherapy. The patients with MAGI3-medium had a robustly good response rate or recurrence-free survival with fluoropyrimidine-based chemotherapy, and were recommended to undergo fluoropyrimidine-based adjuvant chemotherapy. CONCLUSIONS: MAGI3 is a novel E3 ubiquitin ligase by degradation of c-Myc to regulate CRC development and may act as a potential predictor of adjuvant chemotherapy for CRC patients.


Asunto(s)
Neoplasias Colorrectales , Ubiquitina-Proteína Ligasas , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Int J Cancer ; 144(7): 1619-1632, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30230542

RESUMEN

HPV16 is the predominant type of HPV causing invasive cervical cancer. However, the underlying molecular mechanism of the unparalleled carcinogenic power of HPV16 compared to other types of high-risk (HR)-HPV including HPV18 remains elusive. The PDZ binding motif (PBM) of high-risk HPV E6 plays an important role in neoplasia and progression of cervical cancer. HPV16 E6 rather than HPV18 E6, interacted with NHERF1 by its PBM region, and induced degradation of NHERF1. NHERF1 retarded the assembly of cytoskeleton by downregulation of ACTN4, thereby inhibited the migration and invasion of cervical cancer cells in both cell and mouse model. HPV16 E6 was confirmed to enhance actin polymerization with increased ACTN4 level by downregulation of NHERF1, and result in enhanced migration and invasion of cervical cancer cells. GSEA analysis of cervical cancer specimens also showed that HPV16 E6 rather than HPV18 E6, was significantly associated with actin cytoskeleton assembly. That downregulation of NHERF1 by HPV16 E6 promoted cytoskeleton assembly and cell invasion, was an important cause in cervical cancer carcinogenesis. These findings provided the differential mechanism between HPV16 E6 and HPV18 E6 in the development and progression of cervical cancer, which may partially explain the differences of carcinogenic power between these two types of HR-HPVs.


Asunto(s)
Actinina/metabolismo , Papillomavirus Humano 16/patogenicidad , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/metabolismo , Fosfoproteínas/metabolismo , Proteínas Represoras/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Neoplasias del Cuello Uterino/virología , Citoesqueleto de Actina/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/metabolismo , Papillomavirus Humano 18/patogenicidad , Humanos , Ratones , Invasividad Neoplásica , Trasplante de Neoplasias , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/genética , Proteolisis , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
4.
J Cell Mol Med ; 22(2): 1224-1235, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29239102

RESUMEN

Precision therapy for clear cell renal cell carcinoma (ccRCC) requires molecular biomarkers ascertaining disease prognosis. In this study, we performed integrated proteomic and transcriptomic screening in all four tumour-node-metastasis stages of ccRCC and adjacent normal tissues (n = 18) to investigate differentially expressed genes. Most identified differentially expressed genes revealed a strong association with transforming growth factor-ß level and the epithelial-to-mesenchymal transition process. Of them, Serpin peptidase inhibitor clade H member 1 (SERPINH1) revealed the strongest association with poor prognosis and regulation on the expression levels of epithelial-to-mesenchymal transition markers. Subsequently, two independent sets (n = 532 and 105) verified the high level of SERPINH1 in ccRCC tissues and its association with reduced overall survival and disease-free survival in all tumour-node-metastasis stages and patients with von Hippel-Lindau wild-type (VHL-WT). SERPINH1 was an independent predictor of poor overall survival (hazard ratio 0.696 for all patients) and disease-free survival (hazard ratio 0.433 for all patients and 0.362 for patients with VHL-WT) in ccRCC. We have thus shown for the first time that SERPINH1 is an independent precision predictor for unfavourable prognosis in ccRCC. This could assist in identifying patients who need early aggressive management and deepen our understanding of the pathogenesis of VHL-WT ccRCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/metabolismo , Proteínas del Choque Térmico HSP47/metabolismo , Neoplasias Renales/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Mutación/genética , Pronóstico , Proteómica , Reproducibilidad de los Resultados , Transcriptoma/genética , Resultado del Tratamiento , Regulación hacia Arriba , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
5.
FASEB J ; 30(2): 578-89, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26432781

RESUMEN

The actin cytoskeleton is composed of a highly dynamic network of filamentous proteins, yet the molecular mechanism that regulates its organization and remodeling remains elusive. In this study, Na(+)/H(+) exchanger regulatory factor (NHERF)-1 loss-of-function and gain-of-function experiments reveal that polymerized actin cytoskeleton (F-actin) in HeLa cells is disorganized by NHERF1, whereas actin protein expression levels exhibit no detectable change. To elucidate the molecular mechanism underlying actin cytoskeleton disorganization by NHERF1, a combined 2-dimensional electrophoresis-matrix-assisted laser desorption/ionization-time of flight mass spectrometry approach was used to screen for proteins regulated by NHERF1 in HeLa cells. α-Actinin-4, an actin cross-linking protein, was identified. Glutathione S-transferase pull-down and coimmunoprecipitation studies showed the α-actinin-4 carboxyl-terminal region specifically interacted with the NHERF1 postsynaptic density 95/disc-large/zona occludens-1 domain. The NHERF1/α-actinin-4 interaction increased α-actinin-4 ubiquitination and decreased its expression levels, resulting in actin cytoskeleton disassembly. Our study identified α-actinin-4 as a novel NHERF1 interaction partner and provided new insights into the regulatory mechanism of the actin cytoskeleton by NHERF1.


Asunto(s)
Actinina/metabolismo , Citoesqueleto/fisiología , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Actinina/genética , Animales , Células COS , Chlorocebus aethiops , Regulación hacia Abajo , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Fosfoproteínas/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Ubiquitina/metabolismo
6.
Int J Mol Sci ; 18(1)2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28085111

RESUMEN

Cervical cancer is one of the most common female malignancies, and cisplatin-based chemotherapy is routinely utilized in locally advanced cervical cancer patients. However, resistance has been the major limitation. In this study, we found that Na⁺/H⁺ Exchanger Regulatory Factor 1 (NHERF1) was downregulated in cisplatin-resistant cells. Analysis based on a cervical cancer dataset from The Cancer Genome Atlas (TCGA) showed association of NHERF1 expression with disease-free survival of patients received cisplatin treatment. NHERF1 overexpression inhibited proliferation and enhanced apoptosis in cisplatin-resistant HeLa cells, whereas NHERF1 knockdown had inverse effects. While parental HeLa cells were more resistant to cisplatin after NHERF1 knockdown, NHERF1 overexpression in CaSki cells promoted cisplatin sensitivity. Overexpression and knockdown studies also showed that NHERF1 significantly inhibited AKT and extracellular signal-regulated kinase (ERK) signaling pathways in cisplatin-resistant cells. Taken together, our results provide the first evidence that NHERF1 can sensitize cisplatin-refractory cervical cancer cells. This study may help to increase understanding of the molecular mechanisms underlying cisplatin resistance in tumors.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Fosfoproteínas/genética , Intercambiadores de Sodio-Hidrógeno/genética , Neoplasias del Cuello Uterino/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Estimación de Kaplan-Meier , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Fosfoproteínas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Intercambiadores de Sodio-Hidrógeno/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo
7.
BMC Dev Biol ; 15: 51, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26715101

RESUMEN

BACKGROUND: It has been found that microRNAs (miRNAs) play important roles in the regulation of tooth development, and most likely increase the complexity of the genetic network, thus lead to greater complexity of teeth. But there has been no research about the key microRNAs associated with tooth morphogenesis based on miRNAs expression profiles. Compared to mice, the pig model has plentiful types of teeth, which is similar with the human dental pattern. Therefore, we used miniature pigs as large-animal models to investigate differentially expressed miRNAs expression during tooth morphogenesis in the early developmental stages of tooth germ. RESULTS: A custom-designed miRNA microarray with 742 miRNA gene probes was used to analyze the expression profiles of four types of teeth at three stages of tooth development. Of the 591 detectable miRNA transcripts, 212 miRNAs were continuously expressed in all types of tooth germ, but the numbers of miRNA transcript among the four different types of teeth at each embryonic stage were statistically significant differences (p < 0.01). The hierarchical clustering and principal component analysis results suggest that the miRNA expression was globally altered by types and temporal changes. By clustering analysis, we predicted 11 unique miRNA sequences that belong to mir-103 and mir-107, mir-133a and mir-133b, and mir-127 isomiR families. The results of real-time reverse-transcriptase PCR and in situ hybridization experiments revealed that five representative miRNAs may play important roles during different developmental stages of the incisor, canine, biscuspid, and molar, respectively. CONCLUSIONS: The present study indicated that these five miRNAs, including ssc-miR-103 and ssc-miR-107, ssc-miR-133a and ssc-miR-133b, and ssc-miR-127, may play key regulatory roles in different types of teeth during different stages and thus may play critical roles in tooth morphogenesis during early development in miniature pigs.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , MicroARNs/biosíntesis , Morfogénesis/genética , Odontogénesis/genética , Diente/crecimiento & desarrollo , Animales , Dentición , Perfilación de la Expresión Génica , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Componente Principal , Reacción en Cadena en Tiempo Real de la Polimerasa , Sus scrofa
8.
Biochem Biophys Res Commun ; 460(2): 333-40, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25783053

RESUMEN

Ang-(1-7) is an active peptide component of renin-angiotensin system and endogenous ligand for Mas receptor. In the current study, we showed that Ang-(1-7) enhanced migratory and invasive abilities of renal cell carcinoma cells 786-O and Caki-1 by wound-healing, transwell migration and transwell invasion assays. Mas antagonist A779 pretreatment or shRNA-mediated Mas knockdown abolished the stimulatory effect of Ang-(1-7). Furthermore, Ang-(1-7)-stimulated AKT activation was inhibited by either A779 pretreatment or Mas knockdown. Blockage of AKT signaling by AKT inhibitor VIII inhibited Ang-(1-7)-induced migration and invasion in 786-O cells. Taken together, our results provided the first evidence for the pro-metastatic role of Ang-(1-7) in RCC, which may help to better understand the molecular mechanism underlying the progression of this tumor.


Asunto(s)
Angiotensina I/farmacología , Neoplasias Renales/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/metabolismo , Proto-Oncogenes Mas , Cicatrización de Heridas
9.
Amino Acids ; 47(7): 1455-64, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25876703

RESUMEN

The beta-2 adrenergic receptor (ß2AR), a member of GPCR, can activate multiple signaling pathways and is an important treatment target for cardiac failure. However, the molecular mechanism about ß2AR signaling regulation is not fully understood. In this study, we found that cystic fibrosis transmembrane conductance regulator-associated ligand (CAL) overexpression reduced ß2AR-mediated extracellular signal-regulated kinase-1/2 (ERK1/2) activation. Further study identified CAL as a novel binding partner of ß2AR. CAL is associated with ß2AR mainly via the third intracellular loop (ICL3) of receptor and the coiled-coil domains of CAL, which is distinct from CAL/ß1AR interaction mediated by the carboxyl terminal (CT) of ß1AR and PDZ domain of CAL. CAL overexpression retarded ß2AR expression in Golgi apparatus and reduced the receptor expression in plasma membrane.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Receptores Adrenérgicos beta 2/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Aparato de Golgi/metabolismo , Proteínas de la Matriz de Golgi , Células HEK293 , Humanos , Proteínas de Transporte de Membrana , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas
10.
J Nanosci Nanotechnol ; 15(5): 3773-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26505004

RESUMEN

PEGylated hollow mesoporous silica nanoparticles (HMSN-PEG) were successfully fabricated by only one simple step through hydrothermal treatment in Na2CO3 solution. HMSN-PEG nanoparticles were transformed from conventional PEG-modified mesoporous silica nanoparticles (MSN-PEG). The as-synthesized HMSN-PEG nanoparticles exhibited higher loading capacity of anticancer drug (Doxorubicin) and better sustained release property than MSN and MSN-PEG particles. In vitro cell viability of HMSN-PEG nanoparticles to Hep-G2 cells was evaluated. HMSN-PEG nanoparticles have little in vitro cytotoxicity up to a concentration of 500 µg/ml. Furthermore, the DOX-loaded HMSN-PEG nanoparticles exhibited higher cytotoxicity than the DOX-loaded MSN and MSN-PEG nanoparticles against Hep-G2 cells. Therefore, the HMSN-PEG nanoparticle that generated in this PEG protecting etching strategy is a promising nanocarrier toward its potential application for cancer therapy.


Asunto(s)
Portadores de Fármacos/química , Nanopartículas/química , Dióxido de Silicio/química , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Doxorrubicina/farmacología , Portadores de Fármacos/toxicidad , Células Hep G2 , Humanos , Nanopartículas/toxicidad , Porosidad , Dióxido de Silicio/toxicidad
11.
Proc Natl Acad Sci U S A ; 109(33): 13434-9, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22778404

RESUMEN

In vivo recycling of nitrate (NO(3)(-)) and nitrite (NO(2)(-)) is an important alternative pathway for the generation of nitric oxide (NO) and maintenance of systemic nitrate-nitrite-NO balance. More than 25% of the circulating NO(3)(-) is actively removed and secreted by salivary glands. Oral commensal bacteria convert salivary NO(3)(-) to NO(2)(-), which enters circulation and leads to NO generation. The transporters for NO(3)(-) in salivary glands have not yet been identified. Here we report that sialin (SLC17A5), mutations in which cause Salla disease and infantile sialic acid storage disorder (ISSD), functions as an electrogenic 2NO(3)(-)/H(+) cotransporter in the plasma membrane of salivary gland acinar cells. We have identified an extracellular pH-dependent anion current that is carried by NO(3)(-) or sialic acid (SA), but not by Br(-), and is accompanied by intracellular acidification. Both responses were reduced by knockdown of sialin expression and increased by the plasma membrane-targeted sialin mutant (L22A-L23A). Fibroblasts from patients with ISSD displayed reduced SA- and NO(3)(-)-induced currents compared with healthy controls. Furthermore, expression of disease-associated sialin mutants in fibroblasts and salivary gland cells suppressed the H(+)-dependent NO(3)(-) conductance. Importantly, adenovirus-dependent expression of the sialinH183R mutant in vivo in pig salivary glands decreased NO(3)(-) secretion in saliva after intake of a NO(3)(-)-rich diet. Taken together, these data demonstrate that sialin mediates nitrate influx into salivary gland and other cell types. We suggest that the 2NO(3)(-)/H(+) transport function of sialin in salivary glands can contribute significantly to clearance of serum nitrate, as well as nitrate recycling and physiological nitrite-NO homeostasis.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Membrana Celular/metabolismo , Transportadores de Anión Orgánico/metabolismo , Simportadores/metabolismo , Ácidos/metabolismo , Adenoviridae/metabolismo , Animales , Aniones , Transporte Biológico , Fibroblastos/metabolismo , Fibroblastos/patología , Espacio Intracelular/metabolismo , Mutación/genética , Ácido N-Acetilneuramínico/metabolismo , Transportadores de Nitrato , Nitratos/metabolismo , Transportadores de Anión Orgánico/genética , Protones , Enfermedad por Almacenamiento de Ácido Siálico/metabolismo , Glándula Submandibular/citología , Glándula Submandibular/metabolismo , Sus scrofa , Simportadores/genética
12.
BMC Dev Biol ; 14: 16, 2014 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-24750690

RESUMEN

BACKGROUND: The miniature pig provides an excellent experimental model for tooth morphogenesis because its diphyodont and heterodont dentition resembles that of humans. However, little information is available on the process of tooth development or the exact molecular mechanisms controlling tooth development in miniature pigs or humans. Thus, the analysis of gene expression related to each stage of tooth development is very important. RESULTS: In our study, after serial sections were made, the development of the crown of the miniature pigs' mandibular deciduous molar could be divided into five main phases: dental lamina stage (E33-E35), bud stage (E35-E40), cap stage (E40-E50), early bell stage (E50-E60), and late bell stage (E60-E65). Total RNA was isolated from the tooth germ of miniature pig embryos at E35, E45, E50, and E60, and a cDNA library was constructed. Then, we identified cDNA sequences on a large scale screen for cDNA profiles in the developing mandibular deciduous molars (E35, E45, E50, and E60) of miniature pigs using Illumina Solexa deep sequencing. Microarray assay was used to detect the expression of genes. Lastly, through Unigene sequence analysis and cDNA expression pattern analysis at E45 and E60, we found that 12 up-regulated and 15 down-regulated genes during the four periods are highly conserved genes homologous with known Homo sapiens genes. Furthermore, there were 6 down-regulated and 2 up-regulated genes in the miniature pig that were highly homologous to Homo sapiens genes compared with those in the mouse. CONCLUSION: Our results not only identify the specific transcriptome and cDNA profile in developing mandibular deciduous molars of the miniature pig, but also provide useful information for investigating the molecular mechanism of tooth development in the miniature pig.


Asunto(s)
Biblioteca de Genes , Diente Molar/metabolismo , Porcinos Enanos/genética , Diente Primario/metabolismo , Animales , Análisis por Conglomerados , Regulación del Desarrollo de la Expresión Génica , Ontología de Genes , Humanos , Mandíbula/embriología , Mandíbula/metabolismo , Ratones , Diente Molar/embriología , Odontogénesis/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Porcinos Enanos/embriología , Factores de Tiempo , Germen Dentario/embriología , Germen Dentario/metabolismo , Diente Primario/embriología , Transcriptoma/genética
13.
BMC Genomics ; 15: 103, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24498892

RESUMEN

BACKGROUND: Our current knowledge of tooth development derives mainly from studies in mice, which have only one set of non-replaced teeth, compared with the diphyodont dentition in humans. The miniature pig is also diphyodont, making it a valuable alternative model for understanding human tooth development and replacement. However, little is known about gene expression and function during swine odontogenesis. The goal of this study is to undertake the survey of differential gene expression profiling and functional network analysis during morphogenesis of diphyodont dentition in miniature pigs. The identification of genes related to diphyodont development should lead to a better understanding of morphogenetic patterns and the mechanisms of diphyodont replacement in large animal models and humans. RESULTS: The temporal gene expression profiles during early diphyodont development in miniature pigs were detected with the Affymetrix Porcine GeneChip. The gene expression data were further evaluated by ANOVA as well as pathway and STC analyses. A total of 2,053 genes were detected with differential expression. Several signal pathways and 151 genes were then identified through the construction of pathway and signal networks. CONCLUSIONS: The gene expression profiles indicated that spatio-temporal down-regulation patterns of gene expression were predominant; while, both dynamic activation and inhibition of pathways occurred during the morphogenesis of diphyodont dentition. Our study offers a mechanistic framework for understanding dynamic gene regulation of early diphyodont development and provides a molecular basis for studying teeth development, replacement, and regeneration in miniature pigs.


Asunto(s)
Sus scrofa/genética , Diente/metabolismo , Transcriptoma , Animales , Análisis por Conglomerados , Dentición , Redes Reguladoras de Genes , Humanos , Inmunohistoquímica , Morfogénesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Diente/crecimiento & desarrollo
14.
Cell Commun Signal ; 12: 58, 2014 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-25262337

RESUMEN

BACKGROUND: Inositol 1,4,5-trisphosphate receptor (IP3R) plays a pivotal role in the Ca2+ release process in a variety of cell types. Additionally, IP3R is distributed in ventricular intercalated discs, but its function(s) in this particular site remains unknown. Connexin (Cx43), the predominant gap junction (GJ) protein in ventricular myocardium, is linked to several signaling pathways that regulate Cx43 properties by (de)phosphorylation on multiple residues. Here, we investigated the regulatory role of IP3R in cell-cell communication and the mechanism(s) underlying this effect. RESULTS: In neonatal rat and adult mouse ventricular myocytes IP3R co-localized and co-immunoprecipitated with Cx43 in GJ plaques detected by immunostaining and western blot assays. Blocking IP3R with antagonists or silencing pan-IP3R expression with shRNA hindered the 6-carboxyfluorescein (6-CFDA) diffusion through GJs and desynchronized Ca2+ transients among confluent neonatal myocytes in culture, whereas stimulation of IP3R with IP3 ester or ATP exerted the opposite effect. Likewise, 6-CFDA propagation through GJs was modulated by IP3R activation or inhibition in cell pairs of isolated adult cardiomyocytes. Furthermore, IP3R activation or IP3R suppression promoted or suppressed, respectively, Cx43 phosphorylation on S279/282. Site-directed mutagenesis indicated that expression of a mutant Cx43-S282A (alanine) inhibited S279/282 phosphorylation and GJ permeability, while the S279A mutant showed the opposite effect in ventricular myocytes. Expression of these mutants in HEK293 cells revealed that cells with a dual S279/282 mutation failed to express exogenous Cx43, whereas cells with a single S279 or S282 mutation displayed Cx43 overexpression with increased phosphorylation of S279/282 and promotion of intercellular communication. CONCLUSIONS: These results demonstrated, for the first time, that IP3R physically interacts with Cx43 and participates in the regulation of Cx43 phosphorylation on S279/282, thereby affecting GJ intercellular communication in ventricular myocytes.


Asunto(s)
Conexina 43/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Comunicación Celular , Uniones Comunicantes/metabolismo , Uniones Comunicantes/fisiología , Células HEK293 , Ventrículos Cardíacos/citología , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones Endogámicos C57BL , Miocitos Cardíacos/fisiología , Fosforilación , Ratas Sprague-Dawley , Transducción de Señal
15.
Biochem J ; 453(3): 345-56, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23701246

RESUMEN

The functions and signalling mechanisms of the Ang-(1-7) [angiotensin-(1-7)] receptor Mas have been studied extensively. However, less attention has been paid to the intracellular regulation of Mas protein. In the present study, PSD95 (postsynaptic density 95), a novel binding protein of Mas receptor, was identified, and their association was characterized further. Mas specifically interacts with PDZ1-2, but not the PDZ3, domain of PSD95 via Mas-CT (Mas C-terminus), and the last four amino acids [ETVV (Glu-Thr-Val-Val)] of Mas-CT were determined to be essential for this interaction, as shown by GST pull-down, co-immunoprecipitation and confocal co-localization experiments. Gain-of-function and loss-of-function studies indicated that PSD95 enhanced Mas protein expression by increasing the stabilization of the receptor. Mas degradation was robustly inhibited by the proteasome inhibitor MG132 in time- and dose-dependent manners, and the expression of PSD95 impaired Mas ubiquitination, indicating that the PSD95-Mas association inhibits Mas receptor degradation via the ubiquitin-proteasome proteolytic pathway. These findings reveal a novel mechanism of Mas receptor regulation by which its expression is modulated at the post-translational level by ubiquitination, and clarify the role of PSD95, which binds directly to Mas, blocking the ubiquitination and subsequent degradation of the receptor via the ubiquitin-proteasome proteolytic pathway.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Angiotensina I/metabolismo , Animales , Western Blotting , Células COS , Línea Celular , Línea Celular Tumoral , Cricetinae , Homólogo 4 de la Proteína Discs Large , Humanos , Inmunoprecipitación , Fragmentos de Péptidos/metabolismo , Unión Proteica , Proto-Oncogenes Mas , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
16.
J Med Chem ; 67(4): 2802-2811, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38330258

RESUMEN

Chelerythrine chloride (CHE) is a quaternary benzo[c]phenanthridine alkaloid with an iminium group that was found to cause time- and concentration-dependent inhibition of CYP3A4. The loss of CYP3A4 activity was independent of NADPH. CYP3A4 competitive inhibitor ketoconazole and nucleophile N-acetylcysteine (NAC) slowed the inactivation. No recovery of CYP3A4 activity was observed after dialysis. Dihydrochelerythrine hardly inhibited CYP3A4, suggesting that the iminium group was primarily responsible for the inactivation. UV spectral analysis revealed that the maximal absorbance of CHE produced a significant red-shift after being mixed with NAC, suggesting that 1,2-addition possibly took place between the sulfhydryl group of NAC and iminium group of CHE. Molecular dynamics simulation and site-direct mutagenesis studies demonstrated that modification of Cys239 by the iminium group of CHE attributed to the inactivation. In conclusion, CHE is an affinity-labeling inactivator of CYP3A4. The observed enzyme inactivation resulted from the modification of Cys239 of CYP3A4 by the iminium group of CHE.


Asunto(s)
Alcaloides , Antineoplásicos , Benzofenantridinas , Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A/farmacología
17.
Sheng Li Ke Xue Jin Zhan ; 44(6): 403-8, 2013 Dec.
Artículo en Zh | MEDLINE | ID: mdl-24665737

RESUMEN

Epidermal growth factor receptor (EGFR) is imptortant for cell activities, oncogenesis and cell migration, and EGFR inhibitor can treat cancer efficiently, but its side effects, for example, in skin, limited its usage. On the other hand, EGFR interacting proteins may also lead to oncogenesis and its interacting protein as drug targets can avoid cutaneous side effect, which implies possibly a better outcome and life quality of cancer patients. For the multiple EGFR interaction proteins, B1R enhances Erk/MAPK signaling, while PTPN12, Kek1, CEACAM1 and NHERF repress Erk/MAPK signaling. CaM may alter charge of EGFR juxamembrane domain and regulate activation of PI3K/Akt and PLC-gamma/PKC. STAT1, STAT5b are widely thought to be activated by EGFR, while there is unexpectedly inhibiting sequence within EGFR to repress the activity of STATs. LRIG1 and ACK1 enhance the internalization and degration of EGFR, while NHERF and HIP1 repress it. In this article, proteins interacting with EGFR, their interacting sites and their regulation on EGFR signal transduction will be reviewed.


Asunto(s)
Receptores ErbB/fisiología , Transducción de Señal , Movimiento Celular , Humanos , Neoplasias
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 39(2): 103-108, 2023 Feb.
Artículo en Zh | MEDLINE | ID: mdl-36872427

RESUMEN

Objective To establish a Flp-InTM CHO cell line stably expressing human cytochrome P450 oxidoreductase (POR) to lay a solid foundation for the further construction of cell lines stably co-expressing human POR and human cytochrome P450 (CYP). Methods POR recombinant lentivirus was established and infected with Flp-InTM CHO cells, and the expression of green fluorescent protein was observed by fluorescence microscope for monoclonal screening. Mitomycin C (MMC) cytotoxic assay, Western blot analysis and quantitative real-time PCR (qRT-PCR) were employed to detect the activity and expression of POR, and eventually obtained a cell line stably expressing POR (Flp-InTM CHO-POR). Flp-InTM CHO-POR cells (Flp-InTM CHO-POR-2C19) stably co-expressing POR and CYP2C19, and Flp-InTM CHO cells stably expressing CYP2C19 (Flp-InTM CHO-2C19) were constructed, and the activity of CYP2C19 was measured by cyclophosphamide (CPA). Results The consequences of MMC cytotoxic assay, Western blot and qRT-PCR illuminated that Flp-InTM CHO cells infected with POR recombinant lentivirus had elevated MMC metabolic activity and boosted the expression of POR mRNA and protein, compared with Flp-InTM CHO cells infected with negative control virus, indicating that Flp-InTM CHO-POR cells stably expressing POR were obtained. No significant disparity existed in the metabolic activity of CPA between Flp-InTM CHO-2C19 and Flp-InTM CHO cells, whereas the metabolic activity enhanced in Flp-InTM CHO-POR-2C19 and was significantly higher than in Flp-InTM CHO-2C19 cells. Conclusion The stable expression of Flp-InTM CHO-POR cell line is successfully established and can be further applied to construct CYP transgenic cells.


Asunto(s)
Lentivirus , Mitomicina , Animales , Cricetinae , Humanos , Células CHO , Citocromo P-450 CYP2C19 , Cricetulus , Animales Modificados Genéticamente , Western Blotting , Ciclofosfamida
19.
Amino Acids ; 43(5): 2027-35, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22476347

RESUMEN

Ezrin-radixin-moesin-binding phosphoprotein-50 (EBP50) suppresses breast cancer cell proliferation, potentially through its regulatory effect on epidermal growth factor receptor (EGFR) signaling, although the mechanism by which this occurs remains unknown. Thus in our studies, we aimed to determine the effect of EBP50 expression on EGF-induced cell proliferation and activation of EGFR signaling in the breast cancer cell lines, MDA-MB-231 and MCF-7. In MDA-MB-231 cells, which express low levels of EBP50, EBP50 overexpression inhibited EGF-induced cell proliferation, ERK1/2 and AKT phosphorylation. In MCF-7 cells, which express high levels of EBP50, EBP50 knockdown promoted EGF-induced cell proliferation, ERK1/2 and AKT phosphorylation. Knockdown of EBP50 in EBP50-overexpressed MDA-MB-231 cells abrogated the inhibitory effect of EBP50 on EGF-stimulated ERK1/2 phosphorylation and restoration of EBP50 expression in EBP50-knockdown MCF-7 cells rescued the inhibition of EBP50 on EGF-stimulated ERK1/2 phosphorylation, further confirming that the activation of EGF-induced downstream molecules could be specifically inhibited by EBP50 expression. Since EGFR signaling was triggered by EGF ligands via EGFR phosphorylation, we further detected the phosphorylation status of EGFR in the presence or absence of EBP50 expression. Overexpression of EBP50 in MDA-MB-231 cells inhibited EGF-stimulated EGFR phosphorylation, whereas knockdown of EBP50 in MCF-7 cells enhanced EGF-stimulated EGFR phosphorylation. Meanwhile, total expression levels of EGFR were unaffected during EGF stimulation. Taken together, our data shows that EBP50 can suppress EGF-induced proliferation of breast cancer cells by inhibiting EGFR phosphorylation and blocking EGFR downstream signaling in breast cancer cells. These results provide further insight into the molecular mechanism by which EBP50 regulates the development and progression of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fosfoproteínas/genética , Intercambiadores de Sodio-Hidrógeno/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Plásmidos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Transfección
20.
Sci China Life Sci ; 64(11): 1814-1828, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34542810

RESUMEN

Although cisplatin is one of the chemotherapeutics most frequently used in oral squamous cell carcinoma (OSCC) treatment, it exerts multiple side effects and poor chemosensitivity. Nitrate reportedly demonstrates several beneficial biological functions, and synthesized nitrates enhance the therapeutic efficacy of chemotherapy. However, the role of inorganic nitrate in cisplatin chemotherapy remains unclear. We therefore investigated the effect of inorganic nitrate exerted on cisplatin sensitivity in OSCC. We found that nitrate did not affect OSCC cell growth and apoptosis in OSCC cells and OSCC xenograft tumor animal studies. Cisplatin induced REDD1 expression and AKT activation in OSCC. However, nitrate could increase cisplatin chemosensitivity, reduce its REDD1 expression, and attenuate AKT signaling activation in OSCC cells. Dysregulation of high levels of REDD1, which could enhance AKT activation, was positively associated with poor prognosis in OSCC patients. Thus, reduced REDD1 expression and retarded AKT activation induced by inorganic nitrate might be a new potential approach to the sensitization of oral cancer to cisplatin treatment in the future.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias de la Boca/tratamiento farmacológico , Nitratos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA