Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(14): 7288-7313, 2023 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-37378433

RESUMEN

We have conducted a detailed transcriptomic, proteomic and phosphoproteomic analysis of CDK8 and its paralog CDK19, alternative enzymatic components of the kinase module associated with transcriptional Mediator complex and implicated in development and diseases. This analysis was performed using genetic modifications of CDK8 and CDK19, selective CDK8/19 small molecule kinase inhibitors and a potent CDK8/19 PROTAC degrader. CDK8/19 inhibition in cells exposed to serum or to agonists of NFκB or protein kinase C (PKC) reduced the induction of signal-responsive genes, indicating a pleiotropic role of Mediator kinases in signal-induced transcriptional reprogramming. CDK8/19 inhibition under basal conditions initially downregulated a small group of genes, most of which were inducible by serum or PKC stimulation. Prolonged CDK8/19 inhibition or mutagenesis upregulated a larger gene set, along with a post-transcriptional increase in the proteins comprising the core Mediator complex and its kinase module. Regulation of both RNA and protein expression required CDK8/19 kinase activities but both enzymes protected their binding partner cyclin C from proteolytic degradation in a kinase-independent manner. Analysis of isogenic cell populations expressing CDK8, CDK19 or their kinase-inactive mutants revealed that CDK8 and CDK19 have the same qualitative effects on protein phosphorylation and gene expression at the RNA and protein levels, whereas differential effects of CDK8 versus CDK19 knockouts were attributable to quantitative differences in their expression and activity rather than different functions.


Asunto(s)
Quinasas Ciclina-Dependientes , Complejo Mediador , Humanos , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Complejo Mediador/genética , Complejo Mediador/metabolismo , Fosforilación , Proteómica , ARN/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(32): e2201073119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914167

RESUMEN

Breast cancers (BrCas) that overexpress oncogenic tyrosine kinase receptor HER2 are treated with HER2-targeting antibodies (such as trastuzumab) or small-molecule kinase inhibitors (such as lapatinib). However, most patients with metastatic HER2+ BrCa have intrinsic resistance and nearly all eventually become resistant to HER2-targeting therapy. Resistance to HER2-targeting drugs frequently involves transcriptional reprogramming associated with constitutive activation of different signaling pathways. We have investigated the role of CDK8/19 Mediator kinase, a regulator of transcriptional reprogramming, in the response of HER2+ BrCa to HER2-targeting drugs. CDK8 was in the top 1% of all genes ranked by correlation with shorter relapse-free survival among treated HER2+ BrCa patients. Selective CDK8/19 inhibitors (senexin B and SNX631) showed synergistic interactions with lapatinib and trastuzumab in a panel of HER2+ BrCa cell lines, overcoming and preventing resistance to HER2-targeting drugs. The synergistic effects were mediated in part through the PI3K/AKT/mTOR pathway and reduced by PI3K inhibition. Combination of HER2- and CDK8/19-targeting agents inhibited STAT1 and STAT3 phosphorylation at S727 and up-regulated tumor suppressor BTG2. The growth of xenograft tumors formed by lapatinib-sensitive or -resistant HER2+ breast cancer cells was partially inhibited by SNX631 alone and strongly suppressed by the combination of SNX631 and lapatinib, overcoming lapatinib resistance. These effects were associated with decreased tumor cell proliferation and altered recruitment of stromal components to the xenograft tumors. These results suggest potential clinical benefit of combining HER2- and CDK8/19-targeting drugs in the treatment of metastatic HER2+ BrCa.


Asunto(s)
Neoplasias de la Mama , Quinasa 8 Dependiente de Ciclina , Quinasas Ciclina-Dependientes , Resistencia a Antineoplásicos , Inhibidores de Proteínas Quinasas , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quinasa 8 Dependiente de Ciclina/genética , Quinasa 8 Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Lapatinib/farmacología , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor ErbB-2/metabolismo , Trastuzumab/metabolismo , Trastuzumab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Entropy (Basel) ; 25(4)2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-37190468

RESUMEN

In this paper, by using the Hamming distance, we establish a relation between quantum error-correcting codes ((N,K,d+1))s and orthogonal arrays with orthogonal partitions. Therefore, this is a generalization of the relation between quantum error-correcting codes ((N,1,d+1))s and irredundant orthogonal arrays. This relation is used for the construction of pure quantum error-correcting codes. As applications of this method, numerous infinite families of optimal quantum codes can be constructed explicitly such as ((3,s,2))s for all si≥3, ((4,s2,2))s for all si≥5, ((5,s,3))s for all si≥4, ((6,s2,3))s for all si≥5, ((7,s3,3))s for all si≥7, ((8,s2,4))s for all si≥9, ((9,s3,4))s for all si≥11, ((9,s,5))s for all si≥9, ((10,s2,5))s for all si≥11, ((11,s,6))s for all si≥11, and ((12,s2,6))s for all si≥13, where s=s1⋯sn and s1,…,sn are all prime powers. The advantages of our approach over existing methods lie in the facts that these results are not just existence results, but constructive results, the codes constructed are pure, and each basis state of these codes has far less terms. Moreover, the above method developed can be extended to construction of quantum error-correcting codes over mixed alphabets.

4.
Entropy (Basel) ; 24(7)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35885223

RESUMEN

By using difference schemes, orthogonal partitions and a replacement method, some new methods to construct pure quantum error-correcting codes are provided from orthogonal arrays. As an application of these methods, we construct several infinite series of quantum error-correcting codes including some optimal ones. Compared with the existing binary quantum codes, more new codes can be constructed, which have a lower number of terms (i.e., the number of computational basis states) for each of their basis states.

5.
Glycoconj J ; 38(1): 77-87, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33608773

RESUMEN

Liver fibrosis is a continuous wound healing response caused by chronic liver injury, and the activation of hepatic stellate cells (HSCs) is considered as the main event for it. Core fucosylation catalyzed by FUT8 refers to adding the fucosyl moiety to the innermost GlcNAc residue of N-linked oligosaccharides and is involved in many biological processes such as cell differentiation, migration, and signaling transduction. Aberrant core fucosylation is associated with a variety of diseases including cardiovascular disease, tumors and neuroinflammation, but much less is understood in liver fibrosis. Herein, we reported FUT8 mRNA level was increased in patients with liver fibrosis from GEO database and positively correlated with fibrosis progression. FUT8 expression and the core fucosylation were also elevated in TAA-induced mouse liver fibrosis model, and were mainly distributed in the fibrous septum of mouse liver. TGF-ß1, as the most pro-fibrogenic cytokine, could promote the expression of FUT8 and total core fucosylation levels in HSCs in vitro. However, up-regulation of FUT8 in turn inhibited TGF-ß1-induced trans-differentiation, migration and pro-fibrogenic signaling pathways in HSCs. In conclusion, our results suggest that the up-regulation of FUT8 inhibits TGF-ß1-induced HSC activation in a negative feedback loop, and provide potential new therapeutic strategy for liver fibrosis by targeting FUT8.


Asunto(s)
Fucosiltransferasas/genética , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/patología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fucosiltransferasas/metabolismo , Expresión Génica , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Masculino , Ratones Endogámicos C57BL , Ratas , Transducción de Señal/efectos de los fármacos , Tioacetamida/toxicidad , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba
6.
Proc Natl Acad Sci U S A ; 114(38): 10208-10213, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28855340

RESUMEN

The nuclear factor-κB (NFκB) family of transcription factors has been implicated in inflammatory disorders, viral infections, and cancer. Most of the drugs that inhibit NFκB show significant side effects, possibly due to sustained NFκB suppression. Drugs affecting induced, but not basal, NFκB activity may have the potential to provide therapeutic benefit without associated toxicity. NFκB activation by stress-inducible cell cycle inhibitor p21 was shown to be mediated by a p21-stimulated transcription-regulating kinase CDK8. CDK8 and its paralog CDK19, associated with the transcriptional Mediator complex, act as coregulators of several transcription factors implicated in cancer; CDK8/19 inhibitors are entering clinical development. Here we show that CDK8/19 inhibition by different small-molecule kinase inhibitors or shRNAs suppresses the elongation of NFκB-induced transcription when such transcription is activated by p21-independent canonical inducers, such as TNFα. On NFκB activation, CDK8/19 are corecruited with NFκB to the promoters of the responsive genes. Inhibition of CDK8/19 kinase activity suppresses the RNA polymerase II C-terminal domain phosphorylation required for transcriptional elongation, in a gene-specific manner. Genes coregulated by CDK8/19 and NFκB include IL8, CXCL1, and CXCL2, which encode tumor-promoting proinflammatory cytokines. Although it suppressed newly induced NFκB-driven transcription, CDK8/19 inhibition in most cases had no effect on the basal expression of NFκB-regulated genes or promoters; the same selective regulation of newly induced transcription was observed with other transcription signals potentiated by CDK8/19. This selective role of CDK8/19 identifies these kinases as mediators of transcriptional reprogramming, a key aspect of development and differentiation as well as pathological processes.


Asunto(s)
Quinasa 8 Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , FN-kappa B/metabolismo , Quinasa 8 Dependiente de Ciclina/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Citocinas/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos
7.
BMC Cancer ; 18(1): 182, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29439669

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) is a heterogeneous group of hematopoietic malignancies due to sophisticated genetic mutations and epigenetic dysregulation. MicroRNAs (miRNAs), a class of small non-coding RNAs, are important regulators of gene expression in all biological processes, including leukemogenesis. Recently, miR-375 has been reported to be a suppressive miRNA in multiple types of cancers, but its underlying anti-leukemia activity in AML is largely unknown. METHODS: Quantitative reverse transcriptase PCR (qRT-PCR) was used to measure the expression of miR-375 and HOXB3 in leukemic cells and normal controls. Targets of miR-375 were confirmed by western blot and luciferase assay. Phenotypic effects of miR-375 overexpression and HOXB3 knockdown were assessed using viability (trypan blue exclusion assay), colony formation/replating, as well as tumor xenograft assays in vivo. RESULTS: The expression of miR-375 was substantially decreased in leukemic cell lines and primary AML blasts compared with normal controls, because DNA hypermethylation of precursor-miR-375 (pre-miR-375) promoter was discovered in leukemic cells but not in normal controls. Lower expression of miR-375 predicted poor outcome in AML patients. Furthermore, forced expression of miR-375 not only decreased proliferation and colony formation in leukemic cells but also reduced xenograft tumor size and prolonged the survival time in a leukemia xenograft mouse model. Mechanistically, overexpression of miR-375 reduced HOXB3 expression and repressed the activity of a luciferase reporter through binding 3'-untranslated regions (3'-UTR) of HOXB3 mRNA. Overexpression of HOXB3 partially blocked miR-375-induced arrest of proliferation and reduction of colony number, suggesting that HOXB3 plays an important role in miR-375-induced anti-leukemia activity. Knockdown of HOXB3 by short hairpin RNAs reduced the expression of cell division cycle associated 3 (CDCA3), which decreased cell proliferation. Furthermore, HOXB3 induced DNA methyltransferase 3B (DNMT3B) expression to bind in the pre-miR-375 promoter and enhanced DNA hypermethylation of pre-miR-375, leading to the lower expression of miR-375. CONCLUSIONS: Collectively, we have identified a miR-375-HOXB3-CDCA3/DNMT3B regulatory circuitry which contributes to leukemogenesis and suggests a therapeutic strategy of restoring miR-375 expression in AML.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/genética , Leucemia Mieloide/genética , MicroARNs/genética , Regiones no Traducidas 3'/genética , Enfermedad Aguda , Adulto , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Femenino , Células HL-60 , Proteínas de Homeodominio/metabolismo , Humanos , Células K562 , Estimación de Kaplan-Meier , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Persona de Mediana Edad , Trasplante Heterólogo , Adulto Joven , ADN Metiltransferasa 3B
8.
Proc Natl Acad Sci U S A ; 109(34): 13799-804, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869755

RESUMEN

Conventional chemotherapy not only kills tumor cells but also changes gene expression in treatment-damaged tissues, inducing production of multiple tumor-supporting secreted factors. This secretory phenotype was found here to be mediated in part by a damage-inducible cell-cycle inhibitor p21 (CDKN1A). We developed small-molecule compounds that inhibit damage-induced transcription downstream of p21. These compounds were identified as selective inhibitors of a transcription-regulating kinase CDK8 and its isoform CDK19. Remarkably, p21 was found to bind to CDK8 and stimulate its kinase activity. p21 and CDK8 also cooperate in the formation of internucleolar bodies, where both proteins accumulate. A CDK8 inhibitor suppresses damage-induced tumor-promoting paracrine activities of tumor cells and normal fibroblasts and reverses the increase in tumor engraftment and serum mitogenic activity in mice pretreated with a chemotherapeutic drug. The inhibitor also increases the efficacy of chemotherapy against xenografts formed by tumor cell/fibroblast mixtures. Microarray data analysis revealed striking correlations between CDK8 expression and poor survival in breast and ovarian cancers. CDK8 inhibition offers a promising approach to increasing the efficacy of cancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Quinasa 8 Dependiente de Ciclina/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias/tratamiento farmacológico , Animales , Línea Celular Tumoral , Nucléolo Celular/metabolismo , Senescencia Celular , Quinasa 8 Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Genómica , Humanos , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Transcripción Genética , Resultado del Tratamiento
9.
Prostate ; 74(14): 1400-10, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25132524

RESUMEN

BACKGROUND: Gli2, a transcription factor in the Hedgehog pathway, is overexpressed in castrate-resistant prostate cancer (PCa). Previously we showed that Gli2 overexpression increased transcriptional activity of androgen receptor (AR) and conferred androgen growth-independence to normally growth-dependent PCa cells. Here we localized the regions of AR-Gli2 protein interaction and determined the domains within Gli2 needed for AR co-activation. METHODS: Co-immunoprecipitation and GST-pulldown assays were used to define AR-Gli binding domains. Co-activation assays using androgen-responsive promoter reporters were used to define Gli2 regions needed for AR co-activation. Chromatin immunoprecipitation (ChIP) assays were used to confirm nuclear interactions of Gli2 with AR in PCa cells. RESULTS: The Gli2 C-terminal domain (CTD) is sufficient for AR co-activation. Two elements within the CTD were required: (1) an AR binding domain within aa628-897; and (2) at least part of the Gli2 transactivation domain within aa1252-1586. In turn, Gli2 binds the tau5/AF5 ligand-independent activation domain in the AR N-terminus. Mutations in the WxxLF motif in tau5/AF5 greatly diminished binding to Gli2-CTD. Gli2 interaction with AR tau5/AF5 was further substantiated by the ability of Gli2/Gli2-CTD to co-activate truncated AR splice variants (AR-V7/ARV567es). ChIP assays confirmed that Gli2 associates with chromatin at androgen response elements found near androgen-responsive genes in LNCaP cells. These assays also showed that AR associates with chromatin containing a Gli-response element near a Gli-responsive gene. CONCLUSION: Our findings indicate that Gli2 overexpression in PCa cells might support development of castration resistant PCa through AR co-activation and suggests that AR might modulate transcription from Gli2.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Inmunoprecipitación , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Proteínas Nucleares/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Estructura Terciaria de Proteína , Receptores Androgénicos/genética , Elementos de Respuesta , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Transfección , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
10.
Cell Oncol (Dordr) ; 47(1): 113-128, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37578594

RESUMEN

BACKGROUND: Metabolic reprogramming is closely related to the development of gastric cancer (GC), which remains as the fourth leading cause of cancer-related death worldwide. As a tumor suppressor for GC, whether receptor for activated C-kinase 1 (RACK1) play a modulatory role in metabolic reprogramming remains largely unclear. METHODS: GC cell lines and cell-derived xenograft mouse model were used to identify the biological function of RACK1. Flow cytometry and Seahorse assays were applied to examine cell cycle and oxygen consumption rate (OCR), respectively. Western blot, real-time PCR and autophagy double fluorescent assays were utilized to explore the signaling. Immunohistochemistry was performed to detect the expression of RACK1 and other indicators in tissue sections. RESULTS: Loss of RACK1 facilitated the viability, colony formation, cell cycle progression and OCR of GC cells in a glutamine-dependent manner. Further investigation revealed that RACK1 knockdown inhibited the lysosomal degradation of Alanine-serine-cysteine amino acid transporter 2 (ASCT2). Mechanistically, depletion of RACK1 remarkably decreased PTEN expression through up-regulating miR-146b-5p, leading to the activation of AKT/mTOR signaling pathway which dampened autophagy flux subsequently. Moreover, knockdown of ASCT2 could reverse the promotive effect of RACK1 depletion on GC tumor growth both in vitro and in vivo. Tissue microarray confirmed that RACK1 was negatively correlated with the expression of ASCT2 and p62, as well as the phosphorylation of mTOR. CONCLUSION: Together, our results demonstrate that the suppressive function of RACK1 in GC is associated with ASCT2-mediated glutamine metabolism, and imply that targeting RACK1/ASCT2 axis provides potential strategies for GC treatment.


Asunto(s)
Neoplasias Gástricas , Humanos , Animales , Ratones , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Glutamina/metabolismo , Línea Celular Tumoral , Serina-Treonina Quinasas TOR/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
11.
J Clin Invest ; 134(10)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38546787

RESUMEN

Mediator kinases CDK19 and CDK8, pleiotropic regulators of transcriptional reprogramming, are differentially regulated by androgen signaling, but both kinases are upregulated in castration-resistant prostate cancer (CRPC). Genetic or pharmacological inhibition of CDK8 and CDK19 reverses the castration-resistant phenotype and restores the sensitivity of CRPC xenografts to androgen deprivation in vivo. Prolonged CDK8/19 inhibitor treatment combined with castration not only suppressed the growth of CRPC xenografts but also induced tumor regression and cures. Transcriptomic analysis revealed that Mediator kinase inhibition amplified and modulated the effects of castration on gene expression, disrupting CRPC adaptation to androgen deprivation. Mediator kinase inactivation in tumor cells also affected stromal gene expression, indicating that Mediator kinase activity in CRPC molded the tumor microenvironment. The combination of castration and Mediator kinase inhibition downregulated the MYC pathway, and Mediator kinase inhibition suppressed a MYC-driven CRPC tumor model even without castration. CDK8/19 inhibitors showed efficacy in patient-derived xenograft models of CRPC, and a gene signature of Mediator kinase activity correlated with tumor progression and overall survival in clinical samples of metastatic CRPC. These results indicate that Mediator kinases mediated androgen-independent in vivo growth of CRPC, supporting the development of CDK8/19 inhibitors for the treatment of this presently incurable disease.


Asunto(s)
Quinasa 8 Dependiente de Ciclina , Quinasas Ciclina-Dependientes , Neoplasias de la Próstata Resistentes a la Castración , Inhibidores de Proteínas Quinasas , Ensayos Antitumor por Modelo de Xenoinjerto , Masculino , Humanos , Animales , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Ratones , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Quinasa 8 Dependiente de Ciclina/antagonistas & inhibidores , Quinasa 8 Dependiente de Ciclina/genética , Quinasa 8 Dependiente de Ciclina/metabolismo , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
12.
Biol Trace Elem Res ; 201(9): 4508-4517, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36567423

RESUMEN

The objective of this study was to investigate the effects of iron nanoliposomes on iron supplementation and toxicity in SD rats induced by a low-iron diet. The size and infrared spectroscopy of a liposomal oral delivery system were investigated. The particle size of nanoliposomes embedded with chelates was increased. Infrared spectra proved that peptides-iron and blank nanoliposomes were bonded by interaction forces, including the fracture of hydrogen bonds, C = C bonds, hydrophobic interaction, and C-N bonds. We found that iron supplementation chelates had a certain protective effect on viscera after being embedded by nanoliposomes. After 10 days of treatment, the concentration of hemoglobin could be gradually increased. Nanoliposome encapsulated peptides-iron has a better effect than other groups. At the same time, SOD, MDA, and CAT reached normal levels after 20 days. Histological results showed that the sections of the nanoliposomes groups were clearer than those of the other groups. There was a little inflammation in the liver without obvious pathological changes, which also proved that the iron chelates embedded by nanoliposomes had no obvious side effects on iron supplementation in rats. Nanoliposome encapsulated peptides-iron has a small side effect and a significant curative effect of iron supplementation. It maybe has a good application prospect in the clinical medical field.


Asunto(s)
Hierro , Liposomas , Ratas , Animales , Ratas Sprague-Dawley , Liposomas/química , Péptidos/química , Quelantes del Hierro , Suplementos Dietéticos
13.
Prostate ; 72(8): 817-24, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22025366

RESUMEN

Acquired intratumoral steroidogenesis is involved in progression of prostate cancer to castration resistant disease (CRPC) and a target for improved therapeutics. Recent work has shown that prostate cancer cells can acquire steroidogenic activity as they progress to a therapeutic-resistant state. However, benign prostate stromal cells (PrSCs) also have steroidogenic potential though they are often overlooked as a source of intratumoral androgens. Here, we present preliminary studies showing that the steroidogenic activity of primary human PrSCs is significantly increased by exposure to a Hedgehog agonist (SAG) or by transduction of PrSCs with lentiviruses that expresses active Gli2 (Gli2ΔN), a transcription factor that is triggered by Hh signaling. Comparative gene expression profiling on Chips, that was confirmed by quantitative real-time PCR, revealed that hedgehog agonist treatment induced in these cells expressions of hedgehog target genes (Gli1, Ptch1, and SCUBE1) plus a specific cadre of genes involved in cholesterol/steroid biosynthesis, metabolism, and transport. Genes involved downstream in steroid hormone generation, including CYP17A1 and CYP19A1 were also induced. Both the hedgehog agonist and the Gli2-expressing lentivirus significantly increased the output of testosterone (T) from PrSCs that were supplemented with dihydroepiandrosterone (DHEA), an adrenal precursor of T. Finally, knockdown of Gli2 by siRNA suppressed the ability of SAG to induce this response. Collectively, our data indicate that hedgehog/Gli signaling may be a factor in acquired intratumoral steroidogenesis of a prostate tumor through its actions on stromal cells in the tumor microenvironment and an influence for the development of CRPC.


Asunto(s)
Proteínas Hedgehog/fisiología , Proteínas Oncogénicas/fisiología , Comunicación Paracrina/fisiología , Próstata/metabolismo , Esteroides/metabolismo , Células del Estroma/metabolismo , Transactivadores/fisiología , Células Cultivadas , Ciclohexilaminas/farmacología , Deshidroepiandrosterona/metabolismo , Dihidrotestosterona/metabolismo , Proteínas Hedgehog/agonistas , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Próstata/citología , Próstata/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Testosterona/metabolismo , Tiofenos/farmacología , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
14.
Cancer Lett ; 549: 215837, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-35921972

RESUMEN

Metabolic reprogramming is a hallmark in multiple types of malignancies. Fast-growing cancer cells require facilitated synthesis of essential metabolites and excessive energy production. However, whether they are internally coordinated remains largely unknown. Herein, we found that de novo pyrimidine synthesis enhanced aerobic glycolysis in cancer cells. Mechanistically, pyrimidine biosynthesis augmented Notch signaling and transcriptionally increased c-Myc expression, leading to up-regulation of critical glycolytic enzymes. Further studies revealed that pyrimidine synthesis could stabilize γ-secretase subunit Nicastrin at post-translational N-linked glycosylation level, thereby inducing the cleavage and activation of Notch. Besides, we found that up-regulation of the key enzymes for de novo pyrimidine synthesis CAD and DHODH conferred the chemotherapeutic resistance of gastric cancer via accelerating glycolysis, and pharmacologic inhibition of pyrimidine biosynthetic pathway sensitized cancer cells to chemotherapy in vitro and in vivo. Collectively, our findings provide more insights into the regulation of aerobic glycolysis and a metabolic vulnerability that can be exploited to enhance chemotherapy efficacy in gastric cancer.


Asunto(s)
Neoplasias Gástricas , Secretasas de la Proteína Precursora del Amiloide , Resistencia a Antineoplásicos , Glucólisis , Humanos , Pirimidinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico
15.
J Med Chem ; 65(4): 3420-3433, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35114084

RESUMEN

Senexins are potent and selective quinazoline inhibitors of CDK8/19 Mediator kinases. To improve their potency and metabolic stability, quinoline-based derivatives were designed through a structure-guided strategy based on the simulated drug-target docking model of Senexin A and Senexin B. A library of quinoline-Senexin derivatives was synthesized to explore the structure-activity relationship (SAR). An optimized compound 20a (Senexin C) exhibits potent CDK8/19 inhibitory activity with high selectivity. Senexin C is more metabolically stable and provides a more sustained inhibition of CDK8/19-dependent cellular gene expression when compared with the prototype inhibitor Senexin B. In vivo pharmacokinetic (PK) and pharmacodynamic (PD) evaluation using a novel tumor-based PD assay showed good oral bioavailability of Senexin C with a strong tumor-enrichment PK profile and tumor-PD marker responses. Senexin C inhibits MV4-11 leukemia growth in a systemic in vivo model with good tolerability.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Neoplasias del Colon/tratamiento farmacológico , Quinasa 8 Dependiente de Ciclina/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/farmacocinética , Animales , Antineoplásicos/uso terapéutico , Disponibilidad Biológica , Línea Celular Tumoral , Humanos , Leucemia/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/toxicidad , Quinolinas , Relación Estructura-Actividad , Especificidad por Sustrato , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cell Signal ; 83: 110002, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33823241

RESUMEN

Yes-associated protein (YAP) is a vital transcriptional co-activator that activates cell proliferation and evasion of apoptosis for the promotion of tumorigenesis. The von Hippel-Lindau tumor suppressor protein (pVHL), as a critical component of E3 ubiquitin ligase, targets various substrates to regulate tumor progression. However, the precise molecular mechanisms of pVHL during tumorigenesis remain largely unclear. Herein, we found that there was a significant negative correlation between pVHL and YAP at protein level in the TCGA-LUAD dataset and our cohort. Over-expression of pVHL decreased YAP protein expression and reduced its transcriptional activity. Further study indicated that pVHL did not affect YAP mRNA level but decreased YAP protein stability in a lysosome-dependent manner. In addition, the pVHL-mediated degradation of YAP inhibited cellular proliferation, migration, and enhanced chemosensitivity to cisplatin in lung adenocarcinoma cells. Interestingly, the pVHL-mediated YAP degradation was blocked by elevated O-GlcNAcylation. Collectively, our findings demonstrate that pVHL modulates the lysosomal degradation of YAP, and may provide more clues to better understanding the tumor suppressive effects of pVHL.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Carcinogénesis/metabolismo , Neoplasias Pulmonares/metabolismo , Lisosomas/metabolismo , Proteolisis , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Células A549 , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Carcinogénesis/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Lisosomas/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteínas Señalizadoras YAP/genética
17.
Mol Cancer ; 9: 89, 2010 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-20420697

RESUMEN

BACKGROUND: Castration resistant prostate cancer (CRPC) develops as a consequence of hormone therapies used to deplete androgens in advanced prostate cancer patients. CRPC cells are able to grow in a low androgen environment and this is associated with anomalous activity of their endogenous androgen receptor (AR) despite the low systemic androgen levels in the patients. Therefore, the reactivated tumor cell androgen signaling pathway is thought to provide a target for control of CRPC. Previously, we reported that Hedgehog (Hh) signaling was conditionally activated by androgen deprivation in androgen sensitive prostate cancer cells and here we studied the potential for cross-talk between Hh and androgen signaling activities in androgen deprived and androgen independent (AI) prostate cancer cells. RESULTS: Treatment of a variety of androgen-deprived or AI prostate cancer cells with the Hh inhibitor, cyclopamine, resulted in dose-dependent modulation of the expression of genes that are regulated by androgen. The effect of cyclopamine on endogenous androgen-regulated gene expression in androgen deprived and AI prostate cancer cells was consistent with the suppressive effects of cyclopamine on the expression of a reporter gene (luciferase) from two different androgen-dependent promoters. Similarly, reduction of smoothened (Smo) expression with siRNA co-suppressed expression of androgen-inducible KLK2 and KLK3 in androgen deprived cells without affecting the expression of androgen receptor (AR) mRNA or protein. Cyclopamine also prevented the outgrowth of AI cells from androgen growth-dependent parental LNCaP cells and suppressed the growth of an overt AI-LNCaP variant whereas supplemental androgen (R1881) restored growth to the AI cells in the presence of cyclopamine. Conversely, overexpression of Gli1 or Gli2 in LNCaP cells enhanced AR-specific gene expression in the absence of androgen. Overexpressed Gli1/Gli2 also enabled parental LNCaP cells to grow in androgen depleted medium. AR protein co-immunoprecipitates with Gli2 protein from transfected 293T cell lysates. CONCLUSIONS: Collectively, our results indicate that Hh/Gli signaling supports androgen signaling and AI growth in prostate cancer cells in a low androgen environment. The finding that Gli2 co-immunoprecipitates with AR protein suggests that an interaction between these proteins might be the basis for Hedgehog/Gli support of androgen signaling under this condition.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas Hedgehog/metabolismo , Neoplasias de la Próstata/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Andrógenos/genética , Andrógenos/metabolismo , Western Blotting , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Proteínas Hedgehog/genética , Humanos , Inmunoprecipitación , Masculino , Neoplasias de la Próstata/genética , ARN Interferente Pequeño , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Transfección , Alcaloides de Veratrum/farmacología , Proteína con Dedos de Zinc GLI1
18.
J Urol ; 184(1): 344-51, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20488474

RESUMEN

PURPOSE: Hedgehog signaling regulates Gli transcription factors. Aberrant hedgehog signaling can be oncogenic and drugs that block hedgehog are being tested as anticancer agents. We considered whether hedgehog/Gli signaling may be involved in human bladder transitional cell carcinoma proliferative or invasive behavior. MATERIALS AND METHODS: We stratified the human bladder transitional cell carcinoma lines RT4 (ATCC), 253JP, 253BV, UMUC6 and UMUC3 for relative growth rate by cell counting and for in vitro invasiveness by Matrigel invasion assay. Cells were tested for growth inhibition by the hedgehog blocking drug cyclopamine or the inactive mimic tomatidine. Cell RNA was characterized for hedgehog signaling component expression, including ligands, receptors and signaling mediators, by quantitative reverse transcriptase-polymerase chain reaction. Gli2 expression or activity was modified by Gli2 expression lentiviruses or the Gli inhibitor GANT61. We measured effects on proliferation and invasiveness. RESULTS: Cell growth rates and invasiveness were stratified into an equivalent order (RT4 <243JP <253BV

Asunto(s)
Carcinoma de Células Transicionales/genética , Proteínas Hedgehog/genética , Factores de Transcripción de Tipo Kruppel/genética , Invasividad Neoplásica/genética , Proteínas Nucleares/genética , Transducción de Señal/genética , Neoplasias de la Vejiga Urinaria/genética , Western Blotting , Línea Celular Tumoral , Dioxoles/farmacología , Perfilación de la Expresión Génica , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Modelos Lineales , Piperazinas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tomatina/análogos & derivados , Tomatina/farmacología , Alcaloides de Veratrum/farmacología , Proteína Gli2 con Dedos de Zinc
19.
Hum Reprod ; 25(3): 623-32, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20089521

RESUMEN

BACKGROUND: Prostasin is a glycosylphosphatidylinositol-anchored extracellular serine protease with a role in epidermal growth factor receptor (EGFR) signal modulation. EGFR signaling has been shown to be important for regulating cytotrophoblast (CT) cell proliferation in human placenta. We investigated the impact of prostasin expression regulation on this cellular function as well as the molecular mechanisms involved in human cytotrophoblastic cells. METHODS: An immortalized normal human CT cell line (B6Tert-1) was used as an in vitro cell model. Prostasin expression in B6Tert-1 cells was knocked down by transfection of a short interfering RNA. Lentivirus-mediated expression of recombinant human prostasin under tetracycline regulation was performed to obtain stable B6Tert-1 cell sublines that over-expressed prostasin. Changes in cell proliferation and EGFR signaling were evaluated by immunocytochemistry for Ki67 and western blot analysis, respectively, in B6Tert-1 cells with knocked-down or increased prostasin expression. RESULTS: Prostasin knock-down in B6Tert-1 cells resulted in inhibition of cell proliferation, in association with down-regulated EGFR protein expression (both P < 0.05 versus control) as well as reduced phosphorylation of c-raf, mitogen-activated protein kinase (MAPK) kinases (MEK1/2) and extracellular signal-regulated kinases (Erk1/2) (all P < 0.05 versus control). Over-expression of prostasin led to up-regulation of the EGFR protein, but had no effect on cell proliferation or phosphorylation of MAPK signaling molecules in the B6Tert-1 cells. CONCLUSIONS: Prostasin may regulate trophoblast cell proliferation via modulating the EGFR-MAPK signaling pathway.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Receptores ErbB/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Serina Endopeptidasas/fisiología , Trofoblastos/fisiología , Línea Celular , Femenino , Silenciador del Gen , Humanos , Embarazo
20.
Mol Cell Biochem ; 337(1-2): 259-66, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19911255

RESUMEN

The epithelial extracellular serine protease activation cascade involves matriptase (PRSS14) and prostasin (PRSS8), capable of modulating epidermal growth factor receptor (EGFR) signaling. Matriptase activates prostasin by cleaving in the amino-terminal pro-peptide region of prostasin, presumably at the Arg residue of position 44 (R44) of the full-length human prostasin. Using an Arg-to-Ala mutant (R44A) human prostasin, we showed in this report that the cleavage of prostasin by matriptase is at Arg44. This prostasin proteolytic activation site is also cleaved by hepsin (TMPRSS1) to produce active prostasin capable of forming a covalent complex with protease nexin 1 (PN-1). An amino-terminal truncation of EGFR in the extracellular domain (ECD) was observed when the receptor was co-expressed with hepsin. Hepsin and matriptase appear to cleave the EGFR ECD at different sites, while the hepsin cleavage is not affected by active prostasin, which enhances the matriptase cleavage of EGFR. Using hepsin as the prostasin-activating protease in cells co-transfected with EGFR, we showed that active prostasin does not cleave the EGFR ECD directly in the cellular context. Purified active prostasin also does not cleave purified EGFR. Hepsin cleavage of EGFR is not dependent on receptor tyrosine phosphorylation, while the hepsin-cleaved EGFR is phosphorylated at Tyr1068 and no longer responsive to EGF stimulation. The cleavage of EGFR by hepsin does not result in increased phosphorylation of the downstream extracellular signal-regulated kinases (Erk1/2), an event inducible by the matriptase-prostasin cleavage of EGFR. The role of hepsin serine protease should be considered in future studies of epithelial biology concerning matriptase, prostasin, and EGFR.


Asunto(s)
Receptores ErbB/metabolismo , Procesamiento Proteico-Postraduccional , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/fisiología , Células Cultivadas , Activación Enzimática , Epitelio/metabolismo , Epitelio/fisiología , Receptores ErbB/química , Espacio Extracelular , Humanos , Modelos Biológicos , Fosforilación , Estructura Terciaria de Proteína , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA