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










Base de datos
Intervalo de año de publicación
1.
Front Cell Infect Microbiol ; 12: 997018, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36425787

RESUMEN

The incidence of nonalcoholic fatty liver disease (NAFLD) is increasing recently and has become one of the most common clinical liver diseases. Since the pathogenesis of NAFLD has not been completely elucidated, few effective therapeutic drugs are available. As the "second genome" of human body, gut microbiota plays an important role in the digestion, absorption and metabolism of food and drugs. Gut microbiota can act as an important driver to advance the occurrence and development of NAFLD, and to accelerate its progression to cirrhosis and hepatocellular carcinoma. Growing evidence has demonstrated that gut microbiota and its metabolites directly affect intestinal morphology and immune response, resulting in the abnormal activation of inflammation and intestinal endotoxemia; gut dysbiosis also causes dysfunction of gut-liver axis via alteration of bile acid metabolism pathway. Because of its composition diversity and disease-specific expression characteristics, gut microbiota holds strong promise as novel biomarkers and therapeutic targets for NAFLD. Intervening intestinal microbiota, such as antibiotic/probiotic treatment and fecal transplantation, has been a novel strategy for preventing and treating NAFLD. In this article, we have reviewed the emerging functions and association of gut bacterial components in different stages of NAFLD progression and discussed its potential implications in NAFLD diagnosis and therapy.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Humanos , Disbiosis/terapia , Disbiosis/microbiología , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Enfermedad del Hígado Graso no Alcohólico/terapia , Enfermedad del Hígado Graso no Alcohólico/metabolismo
2.
Cell Death Differ ; 28(4): 1251-1269, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33116296

RESUMEN

Inactivation of tumor suppressor Runt-related transcription factor 3 (RUNX3) plays an important role during early tumorigenesis. However, posttranslational modifications (PTM)-based mechanism for the inactivation of RUNX3 under hypoxia is still not fully understood. Here, we demonstrate a mechanism that G9a, lysine-specific methyltransferase (KMT), modulates RUNX3 through PTM under hypoxia. Hypoxia significantly increased G9a protein level and G9a interacted with RUNX3 Runt domain, which led to increased methylation of RUNX3 at K129 and K171. This methylation inactivated transactivation activity of RUNX3 by reducing interactions with CBFß and p300 cofactors, as well as reducing acetylation of RUNX3 by p300, which is involved in nucleocytoplasmic transport by importin-α1. G9a-mediated methylation of RUNX3 under hypoxia promotes cancer cell proliferation by increasing cell cycle or cell division, while suppresses immune response and apoptosis, thereby promoting tumor growth during early tumorigenesis. Our results demonstrate the molecular mechanism of RUNX3 inactivation by G9a-mediated methylation for cell proliferation and antiapoptosis under hypoxia, which can be a therapeutic or preventive target to control tumor growth during early tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Hipoxia de la Célula/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Metilación de ADN/genética , Acetilación , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Phytomedicine ; 78: 153318, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32896707

RESUMEN

BACKGROUND: Hypoxia and HIF-1α are important regulators of tumour growth and angiogenesis and could be attractive targets for cancer therapeutics. Decursin is an active compound extracted from the roots of Angelica gigas and has been shown to have potent anti-cancer and anti-angiogenic activities. However, whether decursin regulates HIF-1α activity and immune responses under hypoxic conditions is not yet understood. PURPOSE: The aim of this study was to identify whether decursin exhibits anti-cancer activity by targeting HIF-1α. STUDY DESIGN: We investigated whether decursin regulates HIF-1α protein stability and increases its degradation. In addition, we determined if decursin increases immune responses in tumour microenvironment to identify its hypoxia-associated anti-cancer activities. MATERIALS AND METHODS: We performed the hypoxia-responsive element promoter-reporter assay, Western blot analysis, immune-fluorescence assay, semi-quantitative RT-PCR and ELISA for VEGF secretion, CCK-8 assay for cell proliferation, TUNEL assay for apoptosis and invasion assay in A549 human lung cancer or HCT116 human colon cancer cells. In vivo Lewis lung carcinoma (LLC) allograft mouse model was used to check tumour growth and immune responses in tumour microenvironment by immunohistochemistry analysis. RESULTS: We observed that decursin inhibited HIF-1 activation under hypoxia by down-regulating the protein level of its subunit HIF-1α. It increased oxygen-dependant hydroxylation and ubiquitination of HIF-1α to promote HIF-1α degradation. Decursin also decreased mRNA expression of HIF-1α target genes. Decursin suppressed cancer cell proliferation, induced apoptosis and inhibited cancer cell invasion under hypoxia in cancer cells. In the allograft mouse tumour model, decursin reduced the hypoxic area and HIF-1α and PD-L1 expression. Infiltrating T cells (CD3+), helper T cells (CD4+) and cytotoxic (CD8+) T cells were accumulated, but regulatory T cells (Foxp3) and myeloid-derived suppressor cell-mediated immune suppressors (Arg1) were attenuated by decursin. CONCLUSION: Our results suggest that decursin is a novel HIF-1α inhibitor that functions by promoting its proteasomal degradation and that it also helps improve T cell activation in tumour microenvironment; these findings provide new explanations about its anti-cancer and anti-angiogenic activity mechanisms.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Benzopiranos/farmacología , Butiratos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Células A549 , Animales , Apoptosis/efectos de los fármacos , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Ratones Endogámicos C57BL , Hipoxia Tumoral/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
BMB Rep ; 53(5): 272-277, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32317087

RESUMEN

Protein kinase CK2 downregulation induces premature senescence in various human cell types via activation of the reactive oxygen species (ROS)-p53-p21Cip1/WAF1 pathway. The transcription factor "nuclear factor erythroid 2-related factor 2" (Nrf2) plays an important role in maintaining intracellular redox homeostasis. In this study, Nrf2 overexpression attenuated CK2 downregulation- induced ROS production and senescence markers including SA-ß-gal staining and activation of p53-p21Cip1/WAF1 in human breast (MCF-7) and colon (HCT116) cancer cells. CK2 downregulation reduced the transcription of Nrf2 target genes, such as glutathione S-transferase, glutathione peroxidase 2, and glutathione reductase 1. Furthermore, CK2 downregulation destabilized Nrf2 protein via inhibiting autophagic degradation of Kelch-like ECHassociated protein 1 (Keap1). Finally, CK2 downregulation decreased the nuclear import of Nrf2 by deactivating AMP-activated protein kinase (AMPK). Collectively, our data suggest that both Keap1 stabilization and AMPK inactivation are associated with decreased activity of Nrf2 in CK2 downregulation-induced cellular senescence. [BMB Reports 2020; 53(5): 272-277].


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Quinasa de la Caseína II/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Humanos , Células Tumorales Cultivadas
5.
J Clin Pharm Ther ; 44(5): 720-725, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31094010

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Teneligliptin is a DPP-4 inhibitor used for the treatment of type 2 diabetes mellitus, commonly prescribed in combination with glimepiride. Teneligliptin is metabolized by CYP3A4, and glimepiride might be partly metabolized by CYP3A4. The aim of the study was to investigate the possible effect of glimepiride on the pharmacokinetics of teneligliptin in healthy subjects. METHODS: A repeated dose, open-label, fixed-sequence study was conducted in 26 healthy subjects. All participants were administered 20 mg teneligliptin daily for 6 days. On day 7, 4 mg glimepiride was administered together with 20 mg teneligliptin. Plasma teneligliptin concentrations were measured at a steady state, and its pharmacokinetic characteristics were compared without and with glimepiride. RESULTS AND DISCUSSION: No statistically significant difference was found in the effect of glimepiride on teneligliptin pharmacokinetics. The steady-state Cmax,ss values of teneligliptin without and with glimepiride were 207.01 ng/mL and 202.15 ng/mL, respectively. Its AUCτ values at steady-state without and with glimepiride were 1527.8 ng · h/mL and 1578.6 ng · h/mL, respectively. The point estimation of geometric mean ratios (GMR) and the 90% confidence interval for both Cmax,ss and AUCτ were within the equivalence range of 0.8-1.25. The results of the present study revealed that glimepiride did not cause pharmacokinetic interaction with teneligliptin in humans. WHAT IS NEW AND CONCLUSION: Glimepiride did not affect the pharmacokinetic characteristics of teneligliptin in healthy subjects.


Asunto(s)
Interacciones Farmacológicas/fisiología , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/uso terapéutico , Pirazoles/farmacocinética , Compuestos de Sulfonilurea/efectos adversos , Compuestos de Sulfonilurea/uso terapéutico , Tiazolidinas/farmacocinética , Adulto , Diabetes Mellitus Tipo 2/metabolismo , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , República de Corea , Adulto Joven
6.
Int J Oncol ; 54(4): 1327-1336, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30968151

RESUMEN

Endothelial progenitor cells (EPCs) are bone marrow (BM)­derived progenitor cells that can differentiate into mature endothelial cells, contributing to vasculogenesis in the blood vessel formation process. Runt­related transcription factor 3 (RUNX3) belongs to the Runt domain family and is required for the differentiation of specific immune cells and neurons. The tumor suppressive role of RUNX3, via the induction of apoptosis and cell cycle arrest in a variety of cancers, and its deletion or frequent silencing by epigenetic mechanisms have been studied extensively; however, its role in the differentiation of EPCs is yet to be investigated. Therefore, in the present study, adult BM­derived hematopoietic stem cells (HSCs) were isolated from Runx3 heterozygous (Rx3+/­) or wild­type (WT) mice. The differentiation of EPCs from the BM­derived HSCs of Rx3+/­ mice was found to be significantly increased compared with those of the WT mice, as determined by the number of small or large colony­forming units. The migration and tube formation abilities of Rx3+/­ EPCs were also observed to be significantly increased compared with those of WT EPCs. Furthermore, the number of circulating EPCs, defined as CD34+/vascular endothelial growth factor receptor 2 (VEGFR2)+ cells, was also significantly increased in Rx3+/­ mice. Hypoxia­inducible factor (HIF)­1α was upregulated in Rx3+/­ EPCs compared with WT EPCs, even under normoxic conditions. Furthermore, in a hindlimb ischemic mouse models, the recovery of blood flow was observed to be highly stimulated in Rx3+/­ mice compared with WT mice. Also, in a Lewis lung carcinoma cell allograft model, the tumor size in Rx3+/­ mice was significantly larger than that in WT mice, and the EPC cell population (CD34+/VEGFR2+ cells) recruited to the tumor was greater in the Rx3+/­ mice compared with the WT mice. In conclusion, the present study revealed that Runx3 inhibits vasculogenesis via the inhibition of EPC differentiation and functions via the suppression of HIF­1α activity.


Asunto(s)
Carcinoma Pulmonar de Lewis/patología , Diferenciación Celular/fisiología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Células Progenitoras Endoteliales/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/patología , Masculino , Ratones , Ratones Transgénicos , Neovascularización Patológica/patología , Cultivo Primario de Células , Regulación hacia Arriba
7.
Int J Oncol ; 54(2): 753-763, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30483760

RESUMEN

The mutation of isocitrate dehydrogenase (IDH)1 (R132H) and IDH2 (R172K) and the induction of hypoxia in various solid tumors results in alterations in metabolic profiles, including the production of the d­ or l­forms of 2­hydroxyglutarate (2HG) from α­ketoglutarate in aerobic metabolism in the tricarboxylic acid (TCA) cycle. However, it is unclear whether the oncometabolite d­2HG increases angiogenesis in endothelial cells. Therefore, in this study, we analyzed the levels of various metabolites, including d­2HG, under hypoxic conditions and in IDH2R172K mutant breast cancer cells by mass spectrometry. We then further evaluated the effects of this metabolite on angiogenesis in breast cancer cells. The results revealed that treatment with d­2HG increased the levels of secreted vascular endothelial growth factor (VEGF) in cancer cells and enhanced endothelial cell proliferation in a concentration­dependent manner. Wound healing and cell migration (examined by Transwell assay) were significantly increased by d­2HG to a level similar to that induced by VEGF. Tube formation was significantly stimulated by d­2HG, and chick chorioallantoic membrane angiogenesis was also enhanced by d­2HG. d­2HG activated VEGF receptor (VEGFR)2 and VEGFR2 downstream signaling, extracellular signal­regulated kinase 1/2, focal adhesion kinase, AKT and matrix metalloproteinase (MMP)2. Taken together, the findings of this study suggested that d­2HG induced angiogenic activity via VEGFR2 signaling and increased MMP2 activity.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Glutaratos/farmacología , Metaloproteinasa 2 de la Matriz/genética , Neovascularización Patológica/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/genética , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Isocitrato Deshidrogenasa/genética , Ácidos Cetoglutáricos/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas c-akt/genética , Hipoxia Tumoral/genética
8.
Exp Mol Med ; 47: e192, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26514084

RESUMEN

Caveolae are flask-shaped cell-surface membranes, which consist of cholesterol, sphingolipids and caveolin proteins. In a microarray analysis, we found that caveolin-1 (Cav-1) was upregulated by receptor activator of NFκB ligand (RANKL), the osteoclast differentiation factor. Silencing of Cav-1 inhibited osteoclastogenesis and also decreased the activation of mitogen-activated protein kinase and the induction of NFATc1 by RANKL. Cav-1 knockdown suppressed the expression of cFms and RANK, two major receptors for osteoclastogenesis. Interestingly, cFms expression was decreased only at the protein level, not at the messenger RNA (mRNA) level, whereas RANK expression was decreased at both the mRNA and protein levels. Furthermore, Cav-1 deficiency increased the lysosomal degradation of cFms. Taken together, these results demonstrate that Cav-1-dependent cFms stabilization contributes to efficient osteoclastogenesis.


Asunto(s)
Caveolina 1/metabolismo , Osteoclastos/citología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Animales , Apoptosis , Caveolina 1/genética , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos ICR , Osteoclastos/metabolismo , Proteolisis , Ligando RANK/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Transducción de Señal
9.
Redox Rep ; 20(5): 228-33, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26018922

RESUMEN

OBJECTIVES: Leukocyte NADPH oxidase, which is active in neutrophils, is a membrane-bound enzyme that catalyzes the reduction of oxygen to O2(-) by using NADPH as an electron donor. Previously, we reported that casein kinase 2 (CK2), a ubiquitous and highly conserved Ser/Thr kinase, is responsible for p47(phox) phosphorylation and that phosphorylation of p47(phox) by CK2 regulates the deactivation of NADPH oxidase. METHODS: Here, we report that the residue Cys(196) of p47(phox) is a target of S-nitrosylation by S-nitrosothiol and peroxynitrite and that this modification enhanced phosphorylation of p47(phox) by CK2. RESULTS: S-Nitrosylated p47(phox) enhanced CK2 b subunit binding, presumably due to alterations in protein conformation. DISCUSSION: Taken together, we propose that S-nitrosylation of p47(phox) regulates the deactivation of NADPH oxidase via enhancement of p47(phox) phosphorylation by CK2.


Asunto(s)
Quinasa de la Caseína II/química , Quinasa de la Caseína II/metabolismo , Quinasa de la Caseína II/genética , Humanos , NADPH Oxidasas/química , NADPH Oxidasas/metabolismo , Fosforilación , Conformación Proteica , S-Nitrosotioles/metabolismo
10.
J Biol Chem ; 290(10): 6522-30, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25561739

RESUMEN

Lipid raft microdomains have important roles in various cellular responses. Caveolae are a specialized type of lipid rafts that are stabilized by oligomers of caveolin proteins. Here, we show that caveolin-1 (Cav-1) plays a crucial role in the regulation of osteoclastogenesis. We found that caveolin-1 was dramatically up-regulated by receptor activator of nuclear factor κB ligand (RANKL), the osteoclast differentiation factor. Knockdown of Cav-1 reduced osteoclastogenesis and induction of NFATc1, the master transcription factor for osteoclastogenesis, by RANKL. Consistent with the in vitro results, injection of caveolin-1 siRNA onto mice calvariae showed reduction in RANKL-induced bone resorption and osteoclast formation. Moreover, Cav-1(-/-) female mice had higher bone volume and lower osteoclast number compared with wild type mice. However, Cav-1(-/-) male mice had both osteoclast and osteoblast numbers higher than wild type mice with no difference in bone volume. The sex dependence in the effect of Cav-1 deficiency was partly attributed to decreased receptor activator of nuclear factor κB and increased cFms expression in osteoclast precursors of female and male mice, respectively. Taken together, these data demonstrate that Cav-1 has a complicated but critical role for osteoclastogenesis.


Asunto(s)
Desarrollo Óseo/genética , Caveolina 1/genética , Diferenciación Celular/genética , Osteoclastos/metabolismo , Animales , Caveolina 1/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Masculino , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Ratones , Factores de Transcripción NFATC/biosíntesis , Osteoclastos/química , Osteogénesis/genética , Ligando RANK/biosíntesis , Caracteres Sexuales
11.
J Biol Chem ; 287(35): 29620-6, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22761415

RESUMEN

Triggering receptor expressed on myeloid cells (TREM)-like transcript-1 (TLT-1) is an immunoreceptor tyrosine-based inhibitory motif (ITIM)-baring TREM family protein. In this study, we identified an alternative transcript form of TLT-1, namely TLT-1s, which has very short extracellular immunoglobulin domain consisting of only 202 amino acids. TLT-1s was mainly expressed in macrophages and osteoclast precursor cells. Upon receptor activator of nuclear factor-κB ligand stimulation, TLT-1s mRNA and protein levels were gradually decreased in BMMs. We also showed the TLT-1s is localized to the cytoplasmic membrane in osteoclast precursor cells. TLT-1s silencing strongly enhanced the formation and resorption activity of osteoclast. In addition, forced expression of TLT-1s showed reduced formation of osteoclast. Because ITIM-baring proteins inhibit immunoreceptor tyrosine-based activation motif (ITAM)-mediated receptor signaling, we tested whether TLT-1s physically interacted with TREM-2, the ITAM-associated co-stimulatory receptor essential for osteoclast differentiation. We showed that TLT-1s is associated with TREM-2 in osteoclast precursor cells. TLT-1s is also associated with tyrosine Src homology 2 domain-containing phosphatase-1 and SH2 domain-containing inositol phosphatase-1 and recruited them to the TREM2-ITAM signaling complex. In addition, knockdown of TLT-1s markedly elevated the intracellular calcium concentration and oscillation in osteoclast precursor cells. In addition, calcium-mediated induction of nuclear factor of activated T cells was also increased by TLT-1s silencing. Furthermore, TREM-2-mediated Akt activation and proliferation of osteoclast precursor cells were also enhanced in TLT-1s silenced cells. In this paper, we found the noble ITIM-baring inhibitory membrane protein; TLT-1s, which regulates ITAM-mediated signaling on osteoclastogenesis.


Asunto(s)
Empalme Alternativo/fisiología , Glicoproteínas de Membrana/metabolismo , Osteoclastos/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal/fisiología , Secuencias de Aminoácidos , Animales , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Activación Enzimática/fisiología , Silenciador del Gen , Glicoproteínas de Membrana/genética , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/citología , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Inmunológicos/genética , Dominios Homologos src
12.
BMB Rep ; 44(9): 572-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21944249

RESUMEN

Elevated phospholipase D (PLD) expression prevents cell cycle arrest and apoptosis. However, the roles of PLD isoforms in cell proliferation and apoptosis are incompletely understood. Here, we investigated the physiological significance of the interaction between PLD2 and protein kinase CKII (CKII) in HCT116 human colorectal carcinoma cells. PLD2 interacted with the CKIIß subunit in HCT116 cells. The C-terminal domain (residues 578-933) of PLD2 and the N-terminal domain of CKIIß were necessary for interaction between the two proteins. PLD2 relocalized CKIIß to the plasma membrane area. Overexpression of PLD2 reduced CKIIß protein level, whereas knockdown of PLD2 led to an increase in CKIIß expression. PLD2-induced CKIIß reduction was mediated by ubiquitin-dependent degradation. The C-terminal domain of PLD2 was sufficient for CKIIß degradation as the catalytic activity of PLD2 was not required. Taken together, the results indicate that the C-terminal domain of PLD2 can regulate CKII by accelerating CKIIß degradation in HCT116 cells.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Neoplasias Colorrectales/metabolismo , Fosfolipasa D/metabolismo , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Células HCT116 , Humanos , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/genética , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ubiquitina/metabolismo
13.
J Bone Miner Res ; 26(6): 1217-29, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21611964

RESUMEN

Nuclear factor of activated T cells c1 (NFATc1) is a transcription factor crucial for the differentiation of osteoclasts. In this study we discovered new signaling pathways involving cAMP regulators that modulate NFATc1 during osteoclastogenesis. The osteoclast differentiation factor receptor activator of NF-κB ligand (RANKL) increased the expression of adenylate cyclase 3 (AC3), accompanied by a rise in the intracellular cAMP level in osteoclasts. The knockdown of AC3 enhanced in vitro osteoclastogenesis and in vivo bone resorption, whereas cAMP-elevating agents showed opposite effects. The antiosteoclastogenic effect of the AC3-cAMP pathway was mediated by the inhibition of NFATc1 nuclear translocation and its autoamplification via a protein kinase A (PKA)-dependent mechanism. RANKL has been shown to activate Ca(2+) /calmodulin-dependent protein kinases (CaMKs). Knockdown or catalytic inhibition of CaMKs elevated intracellular cAMP levels in RANKL-treated osteoclast precursors and suppressed the activation of NFATc1. Taken together, our results demonstrate a pivotal role for the cAMP-PKA-NFATc1 signaling pathway during osteoclast differentiation, suggesting a mechanism by which osteoclastogenesis is fine-tuned by a balance between AC3 and CaMKs activities.


Asunto(s)
Adenilil Ciclasas/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Diferenciación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/enzimología , Osteogénesis , Adenilil Ciclasas/genética , Animales , Resorción Ósea/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , AMP Cíclico/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Modelos Biológicos , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Fosforilación/efectos de los fármacos , Ligando RANK/farmacología , Transducción de Señal/efectos de los fármacos
14.
FEBS Lett ; 583(7): 1164-70, 2009 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-19285079

RESUMEN

Osteoclasts differentiate from macrophage-lineage cells to become specialized for bone resorption function. By a proteomics approach, we found that Lyn was down-regulated by the osteoclast differentiation factor, receptor activator of NF-kappaB ligand (RANKL). The forced reduction of Lyn caused a striking increase in the RANKL-induced PLCgamma1, Ca(2+), and NFATc1 responses during differentiation. These data suggest that Lyn plays a negative role in osteoclastogenesis by interfering with the PLCgamma1-mediated Ca(2+) signaling that leads to NFATc1 activation. Consistent with the in vitro results, in vivo injection of Lyn specific siRNA into mice calvariae provoked a fulminant bone resorption. Our study provides the first evidence of the involvement of Lyn in the negative regulation of osteoclastogenesis by RANKL.


Asunto(s)
Señalización del Calcio/fisiología , Diferenciación Celular/fisiología , Osteoclastos/enzimología , Fosfolipasa C gamma/metabolismo , Ligando RANK/metabolismo , Familia-src Quinasas/biosíntesis , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Humanos , Ratones , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Fosfolipasa C gamma/genética , Proteómica , Ligando RANK/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Cráneo/citología , Cráneo/metabolismo , Familia-src Quinasas/antagonistas & inhibidores
15.
Mol Cells ; 27(3): 299-305, 2009 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19326076

RESUMEN

Over-expression of phospholipase D (PLD) 1 or PLD2 down-regulated CKII activity in NIH3T3 cells. The same results were found with catalytically inactive mutants of PLD isozymes, indicating that the catalytic activity of PLD is not required for PLD-mediated CKII inhibition. Consistent with this, 1-butanol did not alter CKII activity. The reduction in CKII activity in PLD-over-expressing NIH3T3 cells was due to reduced protein level, but not mRNA level, of the CKIIbeta subunit. This PLD-induced CKIIbeta degradation was mediated by ubiquitin-proteasome machinery, but MAP kinase and mTOR were not involved in CKIIbeta degradation. PLD isozymes interacted with the CKIIbeta subunit. Immunocyto-chemical staining revealed that PLD and CKIIbeta colocalize in the cytoplasm of NIH3T3 cells, especially in the perinuclear region. PLD binding to CKIIbeta inhibited CKIIbeta autophosphory-lation, which is known to be important for CKIIbeta stability. In summary, the current data indicate that PLD isozymes can down-regulate CKII activity through the acceleration of CKIIbeta degradation by ubiquitin-proteasome machinery.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Fosfolipasa D/biosíntesis , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/biosíntesis , Quinasa de la Caseína II/genética , Línea Celular Tumoral , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Humanos , Inmunohistoquímica , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Células 3T3 NIH , Fosfolipasa D/genética , Complejo de la Endopetidasa Proteasomal/genética , Transfección
16.
Nat Prod Res ; 22(16): 1441-50, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19023807

RESUMEN

The CKII inhibitory compound was purified from the fruit of Xanthium strumarium by organic solvent extraction and silica gel chromatography. The inhibitory compound was identified as 3,4-dihydroxybenzaldehyde by analysis with FT-IR, FAB-Mass, EI-Mass, (1)H-NMR and (13)C-NMR. 3,4-dihydroxybenzaldehyde inhibited the phosphotransferase activity of CKII with IC(50) of about 783 microM. Steady-state studies revealed that the inhibitor acts as a competitive inhibitor with respect to the substrate ATP. A value of 138.6 microM was obtained for the apparent K(i). Concentration of 300 microM 3,4-dihydroxybenzaldehyde caused 50% growth inhibition of human cancer cell U937. 3,4-dihydroxybenzaldehyde-induced cell death was characterised with the cleavage of poly(ADP-ribose) polymerase and procaspase-3. Furthermore, the inhibitor induced the fragmentation of DNA into multiples of 180 bp, indicating that it triggered apoptosis. This induction of apoptosis by 3,4-dihydroxybenzaldehyde was also confirmed by using flow cytometry analysis. Since CKII is involved in cell proliferation and oncogenesis, these results suggest that 3,4-dihydroxybenzaldehyde may function by inhibiting oncogenic disease, at least in part, through the inhibition of CKII activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzaldehídos/farmacología , Quinasa de la Caseína II/antagonistas & inhibidores , Catecoles/farmacología , Plantas Medicinales/química , Xanthium/química , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Frutas/química , Humanos , Estructura Molecular
17.
Nat Prod Res ; 21(7): 616-24, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17613819

RESUMEN

Cellular DNA topoisomerase I is an important target in cancer chemotherapy. A chloroform extract of the root barks of Cudrania tricuspidata showed an inhibitory effect on mammalian DNA topoisomerase I. The topoisomerase I inhibitory compound was purified and identified as 2',5,7-trihydroxy-4',5'-(2,2-dimethylchromeno)-8-(3-hydroxy-3-methylbutyl) flavanone. The compound, temporarily designated as PKH-3, was shown to inhibit the activity of topoisomerase I with IC50 about 1.0 mM. Concentration of 10 microM PKH-3 caused 50% growth inhibition of human cancer cell U937. PKH-3-induced cell death was characterized with the cleavage of poly(ADP-ribose) polymerase (PARP) and pro-caspase 3. Furthermore, PKH-3 induced the fragmentation of DNA into multiples of 180 b.p. (an apoptotic DNA ladder), indicating that the inhibitor triggered apoptosis. This induction of apoptosis by PKH-3 was also confirmed using flow cytometry analysis. Taken together, these results suggest that PKH-3 may function by inhibiting oncogenic disease, at least in part, through the inhibition of topoisomerase I activity.


Asunto(s)
Antineoplásicos Fitogénicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/aislamiento & purificación , Flavonas/aislamiento & purificación , Flavonas/farmacología , Moraceae/química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Supervivencia Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Flavonas/química , Citometría de Flujo , Humanos , Espectrometría de Masas , Resonancia Magnética Nuclear Biomolecular , Raíces de Plantas/química , Plantas Medicinales/química , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier , Células U937
18.
Gene ; 386(1-2): 147-53, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17069992

RESUMEN

CKII plays a significant role in cell proliferation and cell cycle control. In this report, yeast two-hybrid assay and pull-down assay demonstrate that CKBBP2/CRIF1 associates with the beta subunit of CKII in vitro and in vivo. Recombinant CKBBP2/CRIF1 is phosphorylated in vitro by purified CKII and by CKII inhibitor apigenin-sensitive protein kinase in HEK293 cell extract. Phosphoamino acid analysis and mutational analysis indicate that CKII phosphorylates serine at residue 221 within CKBBP2/CRIF1. Furthermore, serine to alanine mutation at residue 221 abrogates the phosphorylation of CKBBP2/CRIF1 observed in HEK293 cell extract, indicating that CKII is a major kinase that is responsible for phosphorylation of CKBBP2/CRIF1. As compared with the wild-type CKBBP2/CRIF1 or nonphosphorylatable mutant CKBBP2(S221A) (in which the serine-221 is replaced by alanine), overexpression of CKBBP2(S221E) in COS7 cells promotes cell proliferation. Taken together, the present results suggest that CKII may be involved in cell proliferation, at least in part, through the phosphorylation of serine-221 within CKBBP2/CRIF1.


Asunto(s)
Quinasa de la Caseína II/fisiología , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteínas Nucleares/metabolismo , Animales , Células COS , Quinasa de la Caseína II/metabolismo , Chlorocebus aethiops , Células HeLa , Humanos , Fosforilación
19.
Oncol Res ; 15(6): 327-32, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16408697

RESUMEN

The CKII (EC 2.7.1.37) inhibitory compound was purified from the root barks of Cudrania tricuspidata and identified as (2S)-2-[2,4-dihydroxy-5-(3-methyl-but-2-enyl)-phenyl]-5,7-dihyroxy-6-(3-methyl-but-2-enyl)chroman-4-one (euchrestaflavanone B). Euchrestaflavanone B was shown to inhibit the phosphotransferase activity of CKII with IC50 of about 78 microM. Steady-state studies revealed that euchrestaflavanone B acted as a competitive inhibitor with respect to the substrate ATP. A value of 16.4 microM was obtained for the apparent Ki. Concentration of 0.8 microM euchrestaflavanone B caused 50% growth inhibition of human cancer cells U937 and HeLa. Euchrestaflavanone B-induced cell death was characterized with the cleavage of poly(ADP-ribose) polymerase and procaspase-3, indicating that the inhibitor triggered apoptosis. Because protein kinase CKII is involved in cell proliferation and oncogenesis, these results suggest that euchrestaflavanone B may function by inhibiting oncogenic disease, at least in part, through the inhibition of CKII activity.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Quinasa de la Caseína II/antagonistas & inhibidores , Flavanonas/farmacología , Moraceae/química , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Células HeLa , Humanos , Raíces de Plantas/química , Células U937
20.
J Biol Chem ; 278(30): 28079-88, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12716909

RESUMEN

The Gadd45 family of proteins includes Gadd45alpha, MyD118/Gadd45beta, and CR6/OIG37/Gadd45gamma. These proteins play important roles in maintaining genomic stability and in regulating the cell cycle. This study reports the cloning of a novel protein called CR6-interacting factor 1 (CRIF1) which interacts with Gadd45alpha, MyD118/Gadd45beta, and CR6/OIG37/Gadd45gamma. CRIF1 binds specifically to the Gadd45 family proteins, as determined by an in vitro glutathione S-transferase pull-down assay and an in vivo mammalian cell two-hybrid assay along with coimmunoprecipitation assays. CRIF1 mRNA is highly expressed in the thyroid gland, heart, lymph nodes, trachea, and adrenal tissues. CRIF1 localizes exclusively to the nucleus and colocalizes with Gadd45gamma. Recombinant CRIF1 inhibits the histone H1 kinase activity of immunoprecipitated Cdc2-cyclin B1 and Cdk2-cyclin E, and the inhibitory effects were additive with Gadd45 proteins. Overexpression of CRIF1 increases the percentage of cells in G1, decreases the percentage of cells in S phase, and suppresses growth in NIH3T3 cells. The down-regulation of endogenous CRIF1 by the transfection of the small interfering RNA duplexes resulted in the inactivation of Rb by phosphorylation and decreased the G1 phase cell populations. Expression of CRIF1 is barely detectable in adrenal adenoma and papillary thyroid cancer and much lower than in adjacent normal tissue. The results presented here suggest that CRIF1 is a novel nuclear protein that interacts with Gadd45 and may play a role in negative regulation of cell cycle progression and cell growth.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular , Proteínas/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Ciclo Celular , Proteínas de Ciclo Celular/química , División Celular , ADN Complementario/metabolismo , Regulación hacia Abajo , Fase G1 , Glutatión Transferasa/metabolismo , Humanos , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Nucleares , Fosforilación , Pruebas de Precipitina , Unión Proteica , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Proteína de Retinoblastoma/metabolismo , Fase S , Homología de Secuencia de Aminoácido , Factores de Tiempo , Distribución Tisular , Transfección , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos , Proteinas GADD45
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA