Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Carcinog ; 63(5): 991-1008, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38376345

RESUMEN

The worldwide incidence and mortality rates of esophageal squamous cell carcinoma (ESCC) have increased over the last decade. Moreover, molecular targets that may benefit the therapeutics of patients with ESCC have not been fully characterized. Our study discovered that thousand and one amino-acid protein kinase 1 (TAOK1) is highly expressed in ESCC tumor tissues and cell lines. Knock-down of TAOK1 suppresses ESCC cell proliferation in vitro and patient-derived xenograft or cell-derived xenograft tumors growth in vivo. Moreover, TAOK1 overexpression promotes ESCC growth in vitro and in vivo. Additionally, we identified that the natural small molecular compound resveratrol binds to TAOK1 directly and diminishes the kinase activity of TAOK1. Targeting TAOK1 directly with resveratrol significantly inhibits cell proliferation, induces cell cycle arrest and apoptosis, and suppresses tumor growth in ESCC. Furthermore, the silencing of TAOK1 or the application of resveratrol attenuated the activation of TAOK1 downstream signaling effectors. Interestingly, combining resveratrol with paclitaxel, cisplatin, or 5-fluorouracil synergistically enhanced their therapeutic effects against ESCC. In conclusion, this work illustrates the underlying oncogenic function of TAOK1 and provides a theoretical basis for the application of targeting TAOK1 therapy to the clinical treatment of ESCC.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Proteínas Serina-Treonina Quinasas , Humanos , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Resveratrol/farmacología , Resveratrol/uso terapéutico
2.
Biochem Biophys Res Commun ; 545: 150-156, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33550096

RESUMEN

The hypoxia-inducible factor (HIF-1α) functions as a master regulator of oxygen homeostasis. Oxygen-dependent hydroxylation of HIF-1α is tightly regulated by prolyl hydroxylase domain containing proteins (PHD1, PHD2, and PHD3). The prolyl hydroxylation facilitates the recruitment of the von Hippel-Lindau (VHL) protein, leading to ubiquitination and degradation of HIF-1α by the proteasomes. Besides prolyl hydroxylation, phosphorylation of HIF-1α is another central post-translational modification, which regulates its stability under hypoxic conditions as well as normoxic conditions. By use of LC/MS/MS-based analysis, we were able to identify a specific serine residue (Ser451) of HIF-1α phosphorylated under hypoxic conditions. Using plasmids expressing wild type (WT), non-phosphorylatable mutant HIF-1α (S451A), and phosphomimetic mutant HIF-1α (S451E), we demonstrated that the phosphorylation at Ser451 is important in maintaining the HIF-1α protein stability. Notably, phosphorylation at S451 interrupts the interaction of HIF-1α with PHD and pVHL. A phosphomimetic construct of HIF-1α at Ser451 (S451E) is significantly more stable than WT HIF-1α under normoxic conditions. Cells transfected with unphosphorylatable HIF-1α exhibited significantly lower HIF-1 transcriptional activity than WT cells and markedly reduced tumor cell migration. Further, tumors derived from the phosphomimetic mutant cells grew faster, whereas the tumors derived from non-phosphorylatable mutant cells grew slower than the control tumors, suggesting that the phosphorylation of HIF-1α at the Ser451 site is critical to promote tumor growth in vivo. Taken together, our data suggest an alternative mechanism responsible for the regulation of HIF-1α stability.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Sustitución de Aminoácidos , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Hipoxia de la Célula , Células HCT116 , Células HEK293 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Fosforilación , Prolil Hidroxilasas/química , Prolil Hidroxilasas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Serina/química , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/química , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
3.
Biochem Biophys Res Commun ; 533(4): 995-1003, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33012513

RESUMEN

PIN1, the peptidyl-prolyl isomerase (PPIase), is an enzyme that changes the conformation of phosphoproteins. The conformational change induced by PIN1 alters the function and stability of the target proteins. PIN1 is overexpressed in many different types of malignancies, including breast, lung, cervical, brain and colorectal tumors. PIN1 overexpression has been associated with activation of multiple oncogenic signaling pathways during tumor development. Hypoxia-inducible factor 2α (HIF-2α), a transcription factor activated in hypoxia, plays a role in erythropoiesis, glycolysis, tissue invasion, metastasis and angiogenesis. In this study, we found the direct interaction between HIF-2α and PIN1 in colorectal cancer HCT116 cells. Notably, serine 16 and lysine 63 residues of PIN1 were critical for its interaction with HIF-2α. When PIN1 protein was silenced by transient transfection of PIN1 short interfering RNA, the expression of HIF-2α was attenuated under a hypoxic condition. Moreover, genetic and pharmacologic inhibition of PIN1 abrogated the expression of vascular endothelial growth factor and angiogenesis. The cycloheximide chase experiment revealed the stabilization of HIF-2α by PIN1. Both WW and PPIase domains of PIN1 appear to be critical for its interaction with HIF-2α.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Neovascularización Patológica/etiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Embrión de Pollo , Femenino , Células HCT116 , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Células 3T3 NIH , Peptidilprolil Isomerasa de Interacción con NIMA/química , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Interferencia de ARN , ARN Interferente Pequeño/genética , Hipoxia Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Arch Biochem Biophys ; 689: 108413, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32473133

RESUMEN

The proviral integration site for Moloney murine leukemia virus (PIM) family of serine/threonine-specific kinases consist of three isoforms, that regulate proliferation, apoptosis, metabolism, invasion, and metastasis of cancer cells. Among these, abnormally elevated kinase activity of PIM-1 contributes to the progression of gastric cancer and predicts poor prognosis and a low survival rate in gastric cancer patients. In the present study, we found that resveratrol, one of the representative chemopreventive and anticarcinogenic phytochemicals, directly binds to PIM-1 and thereby inhibits its catalytic activity in human gastric cancer SNU-601 cells. This resulted in suppression of phosphorylation of the proapoptotic Bad, a known substrate of PIM-1. Resveratrol, by inactivating PIM-1, also inhibited anchorage-independent growth and proliferation of SNU-601 cells. To understand the molecular interaction between resveratrol and PIM-1, we conducted docking simulation and found that resveratrol directly binds to the PIM-1 at the ATP-binding pocket. In conclusion, the proapototic and anti-proliferative effects of resveratrol in gastric cancer cells are likely to be mediated through suppression of PIM-1 kinase activity, which may represent a novel mechanism underlying its chemopreventive and anticarcinogenic actions.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Resveratrol/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Neoplasias Gástricas/metabolismo
5.
Int J Mol Sci ; 21(14)2020 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-32664659

RESUMEN

Persistent hair loss is a major cause of psychological distress and compromised quality of life in millions of people worldwide. Remarkable progress has been made in understanding the molecular basis of hair loss and identifying valid intracellular targets for designing effective therapies for hair loss treatment. Whereas a variety of growth factors and signaling pathways have been implicated in hair cycling process, the activation of Wnt/ß-catenin signaling plays a central role in hair follicle regeneration. Several plant-derived chemicals have been reported to promote hair growth by activating Wnt/ß-catenin signaling in various in vitro and in vivo studies. This mini-review sheds light on the role of Wnt/ß-catenin in promoting hair growth and the current progress in designing hair loss therapies by targeting this signaling pathway.


Asunto(s)
Alopecia/terapia , Preparaciones para el Cabello/uso terapéutico , Trasplante de Células Madre Mesenquimatosas , Terapia Molecular Dirigida , Vía de Señalización Wnt/efectos de los fármacos , Alopecia/tratamiento farmacológico , Alopecia/metabolismo , Animales , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Terapia Combinada , Femenino , Cabello/crecimiento & desarrollo , Folículo Piloso/efectos de los fármacos , Folículo Piloso/fisiología , Preparaciones para el Cabello/farmacología , Humanos , Masculino , Ratones , Ratones Desnudos , Fitoterapia , Regeneración/efectos de los fármacos
6.
Int J Mol Sci ; 20(12)2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-31208018

RESUMEN

Sesquiterpene lactones constitute a major class of bioactive natural products. One of the naturally occurring sesquiterpene lactones is costunolide, which has been extensively investigated for a wide range of biological activities. Multiple lines of preclinical studies have reported that the compound possesses antioxidative, anti-inflammatory, antiallergic, bone remodeling, neuroprotective, hair growth promoting, anticancer, and antidiabetic properties. Many of these bioactivities are supported by mechanistic details, such as the modulation of various intracellular signaling pathways involved in precipitating tissue inflammation, tumor growth and progression, bone loss, and neurodegeneration. The key molecular targets of costunolide include, but are not limited to, intracellular kinases, such as mitogen-activated protein kinases, Akt kinase, telomerase, cyclins and cyclin-dependent kinases, and redox-regulated transcription factors, such as nuclear factor-kappaB, signal transducer and activator of transcription, activator protein-1. The compound also diminished the production and/expression of proinflammatory mediators, such as cyclooxygenase-2, inducible nitric oxide synthase, nitric oxide, prostaglandins, and cytokines. This review provides an overview of the therapeutic potential of costunolide in the management of various diseases and their underlying mechanisms.


Asunto(s)
Fitoquímicos/farmacología , Sesquiterpenos/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Antioxidantes/química , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Humanos , Fitoquímicos/química , Fitoquímicos/uso terapéutico , Sesquiterpenos/química , Sesquiterpenos/uso terapéutico
7.
Int J Mol Sci ; 20(2)2019 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-30646538

RESUMEN

The hYSK1, a serine/threonine kinase (STK)-25, has been implicated in a variety of cellular functions including cell migration and polarity. We have recently reported that hYSK1 down-regulated the expression and functions of p16INK4a, a cell cycle regulatory protein, thereby enhancing migration and growth of cancer cells under hypoxic conditions. In this study, we further investigated the mechanisms underlying downregulation of p16INK4a and anti-migratory function of hYSK1. Our study revealed that p21WAF1/Cip1 is a novel binding partner of hYSK1. Moreover, the interaction between hYSK1 and p21WAF1/Cip1 led to the inhibition of SP-1 transcriptional activity, as revealed by a significant down-regulation of SP-1-mediated transactivation of p16INK4a promoter, and accelerated MMP-2 expression. Conversely, the knock-down of hYSK1 enhanced the p16INK4a promoter activity and protein expression, and diminished MMP-2 transcription and protein levels in hypoxic conditions as compared to control. Taken together, hYSK1 blocks the p21WAF1/Cip1 functions by direct interaction and inhibits the p16INK4a expression and induces MMP-2 expression by its regulations of SP-1 transcriptional activity under the hypoxia conditions.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Serina-Treonina Quinasas/genética , Transcripción Genética , Hipoxia de la Célula/genética , Línea Celular , Movimiento Celular/genética , Polaridad Celular/genética , Regulación de la Expresión Génica , Humanos , Metaloproteinasa 2 de la Matriz/genética , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas , Unión Proteica , Mapas de Interacción de Proteínas/genética
8.
Molecules ; 24(2)2019 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-30642127

RESUMEN

Apple is a rich source of bioactive phytochemicals that help improve health by preventing and/or curing many disease processes, including cancer. One of the apple polyphenols is phloretin [2',4',6'-Trihydroxy-3-(4-hydroxyphenyl)-propiophenone], which has been widely investigated for its antioxidant, anti-inflammatory and anti-cancer activities in a wide array of preclinical studies. The efficacy of phloretin in suppressing xenograft tumor growth in athymic nude mice implanted with a variety of human cancer cells, and the ability of the compound to interfere with cancer cells signaling, have made it a promising candidate for anti-cancer drug development. Mechanistically, phloretin has been reported to arrest the growth of tumor cells by blocking cyclins and cyclin-dependent kinases and induce apoptosis by activating mitochondria-mediated cell death. The blockade of the glycolytic pathway via downregulation of GLUT2 mRNA and proteins, and the inhibition of tumor cells migration, also corroborates the anti-cancer effects of phloretin. This review sheds light on the molecular targets of phloretin as a potential anti-cancer and anti-inflammatory natural agent.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Chalconas/química , Chalconas/farmacología , Floretina/química , Floretina/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Humanos , Modelos Animales
9.
Int J Mol Sci ; 19(9)2018 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-30208587

RESUMEN

The functional aspect of scalp hair is not only to protect from solar radiation and heat/cold exposure but also to contribute to one's appearance and personality. Progressive hair loss has a cosmetic and social impact. Hair undergoes three stages of hair cycle: the anagen, catagen, and telogen phases. Through cyclical loss and new-hair growth, the number of hairs remains relatively constant. A variety of factors, such as hormones, nutritional status, and exposure to radiations, environmental toxicants, and medications, may affect hair growth. Androgens are the most important of these factors that cause androgenic alopecia. Other forms of hair loss include immunogenic hair loss, that is, alopecia areata. Although a number of therapies, such as finasteride and minoxidil, are approved medications, and a few others (e.g., tofacitinib) are in progress, a wide variety of structurally diverse classes of phytochemicals, including those present in ginseng, have demonstrated hair growth-promoting effects in a large number of preclinical studies. The purpose of this review is to focus on the potential of ginseng and its metabolites on the prevention of hair loss and its underlying mechanisms.


Asunto(s)
Alopecia/tratamiento farmacológico , Cabello/efectos de los fármacos , Cabello/crecimiento & desarrollo , Panax , Fitoterapia , Preparaciones de Plantas/uso terapéutico , Alopecia/metabolismo , Alopecia/prevención & control , Animales , Cabello/metabolismo , Folículo Piloso/efectos de los fármacos , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/metabolismo , Humanos , Panax/química , Panax/metabolismo , Fitoterapia/métodos , Preparaciones de Plantas/química , Preparaciones de Plantas/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Int J Mol Sci ; 17(3): 290, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26959007

RESUMEN

Cancer, being the second leading cause of mortality, exists as a formidable health challenge. In spite of our enormous efforts, the emerging complexities in the molecular nature of disease progression limit the real success in finding an effective cancer cure. It is now conceivable that cancer is, in fact, a progressive illness, and the morbidity and mortality from cancer can be reduced by interfering with various oncogenic signaling pathways. A wide variety of structurally diverse classes of bioactive phytochemicals have been shown to exert anticancer effects in a large number of preclinical studies. Multiple lines of evidence suggest that withaferin-A can prevent the development of cancers of various histotypes. Accumulating data from different rodent models and cell culture experiments have revealed that withaferin-A suppresses experimentally induced carcinogenesis, largely by virtue of its potent anti-oxidative, anti-inflammatory, anti-proliferative and apoptosis-inducing properties. Moreover, withaferin-A sensitizes resistant cancer cells to existing chemotherapeutic agents. The purpose of this review is to highlight the mechanistic aspects underlying anticancer effects of withaferin-A.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias/tratamiento farmacológico , Witanólidos/farmacología , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Anticarcinógenos/química , Anticarcinógenos/farmacología , Anticarcinógenos/uso terapéutico , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Neoplasias/irrigación sanguínea , Neoplasias/patología , Withania/química , Witanólidos/química , Witanólidos/uso terapéutico
11.
J Cell Sci ; 126(Pt 8): 1744-52, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23444377

RESUMEN

The tumor suppressor protein p16(INK4a) is a member of the INK4 family of cyclin-dependent kinase (Cdk) inhibitors, which are involved in the regulation of the eukaryotic cell cycle. However, the mechanisms underlying the anti-proliferative effects of p16(INK4a) have not been fully elucidated. Using yeast two-hybrid screening, we identified the eukaryotic elongation factor (eEF)1A2 as a novel interacting partner of p16(INK4a). eEF1A2 is thought to function as an oncogene in cancers. The p16(INK4a) protein interacted with all but the D2 (250-327 aa) domain of eEF1A2. Ectopic expression of p16(INK4a) decreased the expression of eEF1A2 and inhibited cancer cell growth. Furthermore, suppression of protein synthesis by expression of p16(INK4a) ex vivo was verified by luciferase reporter activity. Microinjection of p16(INK4a) mRNA into the cytoplasm of Xenopus embryos suppressed the luciferase mRNA translation, whereas the combination of p16(INK4a) and morpholino-eEF1A2 resulted in a further reduction in translational activity. We conclude that the interaction of p16(INK4a) with eEF1A2, and subsequent downregulation of the expression and function of eEF1A2 is a novel mechanism explaining the anti-proliferative effects of p16(INK4a).


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Factor 1 de Elongación Peptídica/metabolismo , Animales , Western Blotting , Células CHO , Células COS , Línea Celular Tumoral , Proliferación Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Células HeLa , Humanos , Inmunoprecipitación , Unión Proteica , Técnicas del Sistema de Dos Híbridos , Xenopus laevis
12.
Bioorg Med Chem Lett ; 25(23): 5625-31, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26508549

RESUMEN

Somatic heterozygous mutations of isocitrate dehydrogenase-1 (IDH1) are abundantly found in several types of cancer and strongly implicate altered metabolism in carcinogenesis. In the present study, we have identified α-mangostin as a novel selective inhibitor of mutant IDH1 (IDH1-R132H). We have observed that α-mangostin competitively inhibits the binding of α-ketoglutarate (α-KG) to IDH1-R132H. The structure-relationship study reveals that α-mangostin exhibits the strongest core inhibitor structure. Finally, we have observed that α-mangostin selectively promotes demethylation of 5-methylcytosine (5mC) and histone H3 trimethylated lysine residues in IDH1 (+/R132H) MCF10A cells, presumably via restoring the activity of cellular α-KG-dependent DNA hydroxylases and histone H3 lysine demethylases. Collectively, we provide evidence that α-mangostin selectively inhibits IDH1-R132H.


Asunto(s)
Descubrimiento de Drogas , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Xantonas/química , Unión Competitiva , Humanos , Isocitrato Deshidrogenasa/genética , Células MCF-7 , Estructura Molecular , Mutación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Relación Estructura-Actividad , Xantonas/farmacología
13.
Bioorg Med Chem Lett ; 24(19): 4763-4767, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25205188

RESUMEN

Natural compounds are regarded as a rich source for potential anti-inflammatory and anti-carcinogenic agents. Increasing evidence indicates that histone phosphorylation at Ser10 is a marker for cell cycle progression during the mitosis and the induction of immediate pro-inflammatory genes during the interphase. In the present study, we have screened our in-house natural compounds to find out new chemical inhibitor(s) of histone H3 phosphorylation at Ser10. As a result, we observed that α-amyrin, oleanolic acid, marliolide, and 4'-O-ß-D-glucosyl-5-O-methylvisamminol decreased the levels of histone H3 phosphorylation at Ser10 and c-Jun. In particular, we observed that 4'-O-ß-D-glucosyl-5-O-methylvisamminol suppressed the direct interaction of histone H3 with 14-3-3ε, inhibited the aurora B kinase activity and delayed the mitotic cell cycle progression. We reports 4'-O-ß-D-glucosyl-5-O-methylvisamminol as the first epigenetic natural chemical inhibitor that can abrogates the mitotic cell cycle progression and immediate pro-inflammatory gene expressions via suppression of histone H3 phosphorylation at Ser10 and its interaction with 14-3-3ε.


Asunto(s)
Proteínas 14-3-3/antagonistas & inhibidores , Cromonas/farmacología , Epigénesis Genética/efectos de los fármacos , Glucósidos/farmacología , Histonas/antagonistas & inhibidores , Serina/antagonistas & inhibidores , Proteínas 14-3-3/metabolismo , Supervivencia Celular/efectos de los fármacos , Cromonas/síntesis química , Cromonas/química , Glucósidos/síntesis química , Glucósidos/química , Células HT29 , Histonas/metabolismo , Humanos , Conformación Molecular , Fosforilación/efectos de los fármacos , Serina/metabolismo , Relación Estructura-Actividad
14.
Molecules ; 19(7): 10074-89, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25014534

RESUMEN

Extracellular and intracellular oxidants or electrophiles are key contributors to the damages in cellular macromolecules, such as DNA, proteins and lipids. Nrf2 is a master transcription factor that modulates a cellular antioxidant response program and plays an important role in the protection against oxidants and electrophiles. Keap1 is a regulator of Nrf2 by serving as a substrate adaptor for Cullin3-dependent E3 ubiquitin ligase. While Nrf2 activation is a feasible strategy for treatment of age-related diseases, aberrant Nrf2 activation also confers a selective growth advantage of tumor cells during chemotherapy or radiotherapy. In the present review, we provide an overview of the Keap1-Nrf2-ARE system, the domain organization of Nrf2 and Keap1, and the regulatory mechanisms of Nrf2 proteolysis by Keap1. We also discuss how Nrf2 prevents tumor promotion, hampers the sensitivity of selected tumors against chemotherapy or radiotherapy, and reprograms the metabolism to facilitate the tumor proliferation. Finally, we illustrate the current status in the development of Nrf2 chemical activators and inhibitors for the use of potential chemopreventive agents and chemotherapeutic adjuvants, respectively.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal , Animales , Descubrimiento de Drogas , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Factor 2 Relacionado con NF-E2/agonistas , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/química , Factor 2 Relacionado con NF-E2/genética , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal/efectos de los fármacos
15.
J Cosmet Dermatol ; 21(7): 2763-2773, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35596731

RESUMEN

BACKGROUND: Lagerstroemia indica (L. indica) is reported to have diverse biological activities including anti-inflammatory, anti-cancer, neuro-regulatory, antidiabetic, and antioxidant activity. AIMS: The purpose of this study is to examine the potential of hair growth promotion and/or hair loss prevention by L. indica extract. PATIENTS/METHODS: The effects of L. indica on hair growth have been studied in human hair follicle dermal papillary (hHFDP) cells and follicular organ culture ex vivo by cell proliferation assay, PCR, western blot analysis, and reporter gene activity assay. Moreover, a clinical trial was conducted in healthy volunteers. RESULTS: Lagerstroemia indica significantly promoted the proliferation of hHFDP cells, which was associated with increased expression of TCF/LEF, VEGF, and Gli1 mRNA, and inhibition of STAT6 and Smad2 mRNA. Treatment with L. indica also increased the TCF/LEF reporter gene activity but downregulated the SBE- and STAT6-luciferase activities. The expression of total ß-catenin, CDK4, and CDK2 were elevated, while that of STAT6 and SMAD2/3 was suppressed upon treatment with L. indica. In human hair follicles organ culture, L. indica significantly inhibited hair follicular degeneration. The clinical trial showed a statistically significant rise in total hair count in test group (n = 24) after 24 weeks of applying the hair tonic enriched with L. indica (141.46 ± 21.27 number/cm2 , p < 0.05). CONCLUSION: We suggest that L. indica extract prevents hair loss as well as stimulate hair growth by regulating the Wnt-ß-catenin, JAK3-STAT6, and TGF-ß1-Smad signaling pathways, and may be further developed as a novel functional cosmetic for preventing hair loss.


Asunto(s)
Lagerstroemia , beta Catenina , Alopecia/metabolismo , Proliferación Celular , Células Cultivadas , Cabello , Folículo Piloso , Humanos , Lagerstroemia/genética , Lagerstroemia/metabolismo , Extractos Vegetales/metabolismo , Extractos Vegetales/farmacología , ARN Mensajero/metabolismo , Factor de Transcripción STAT6/metabolismo , Factor de Transcripción STAT6/farmacología , Vía de Señalización Wnt , beta Catenina/metabolismo
16.
FEBS Lett ; 595(5): 604-622, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33452674

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) has been considered as a potential target for development of anticancer therapeutics. Here, we report a novel mechanism by which the cyclopentenone prostaglandin, 15-deoxy-Δ12,14 -prostaglandin J2 (15d-PGJ2 ) functions as an allosteric inhibitor of STAT3. 15d-PGJ2 inhibits phosphorylation, dimerization, nuclear translocation, and transcriptional activity of STAT3 in H-Ras-transformed human mammary epithelial cells (MCF10A-Ras) through the Michael addition reaction at cysteine 259 of STAT3. Comparative studies with 15d-PGJ2 analogues reveal that both C12-C13 and C9-C10 double bonds conjugated to the carbonyl group in the cyclopentenone ring of 15d-PGJ2 are essential for STAT3 binding. Antiproliferative and pro-apoptotic activities of 15d-PGJ2 in MCF10A-Ras cells are attributable to covalent modification of STAT3 on Cys259, and mimic the effects induced by mutation of this amino acid.


Asunto(s)
Antineoplásicos/farmacología , Cisteína/química , Células Epiteliales/efectos de los fármacos , Prostaglandina D2/análogos & derivados , Proteínas Proto-Oncogénicas p21(ras)/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Secuencia de Aminoácidos , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Cisteína/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Fosforilación/efectos de los fármacos , Prostaglandina D2/química , Prostaglandina D2/farmacología , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Relación Estructura-Actividad , Transcripción Genética
17.
Front Cell Dev Biol ; 9: 638174, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33842463

RESUMEN

BACKGROUND: Skin cancer is one of the most commonly diagnosed cancers worldwide. The 5-year survival rate of the most aggressive late-stage skin cancer ranges between 20 and 30%. Thus, the discovery and investigation of novel target therapeutic agents that can effectively treat skin cancer is of the utmost importance. The T-lymphokine-activated killer cell-originated protein kinase (TOPK), which belongs to the serine-threonine kinase class of the mitogen-activated protein kinase kinase (MAPKK) family, is highly expressed and activated in skin cancer. The present study investigates the role of 3-deoxysappanchalcone (3-DSC), a plant-derived functional TOPK inhibitor, in suppressing skin cancer cell growth. PURPOSE: In the context of skin cancer prevention and therapy, we clarify the effect and mechanism of 3-DSC on different types of skin cancer and solar-simulated light (SSL)-induced skin hyperplasia. METHODS: In an in vitro study, western blotting and in vitro kinase assays were utilized to determine the protein expression of TOPK and its activity, respectively. Pull-down assay with 3-DSC and TOPK (wild-type and T42A/N172 mutation) was performed to confirm the direct interaction between T42A/N172 amino acid sites of TOPK and 3-DSC. Cell proliferation and anchorage-independent cell growth assays were utilized to determine the effect of 3-DSC on cell growth. In an in vivo study, the thickness of skin and tumor size were measured in the acute SSL-induced inflammation mouse model or SK-MEL-2 cell-derived xenografts mouse model treated with 3-DSC. Immunohistochemistry analysis of tumors isolated from SK-MEL-2 cell-derived xenografts was performed to determine whether cell-based results observed upon 3-DSC treatment could be recapitulated in vivo. RESULTS: 3-DSC is able to inhibit cell proliferation in skin cancer cells in an anchorage-dependent and anchorage-independent manner by regulation of TOPK and its related signaling pathway in vitro. We also found that application of 3-DSC reduced acute SSL-induced murine skin hyperplasia. Additionally, we observed that 3-DSC decreased SK-MEL-2 cell-derived xenograft tumor growth through attenuating phosphorylation of TOPK and its downstream effectors including ERK, RSK, and c-Jun. CONCLUSIONS: Our results suggest that 3-DSC may function in a chemopreventive and chemotherapeutic capacity by protecting against UV-induced skin hyperplasia and inhibiting tumor cell growth by attenuating TOPK signaling, respectively.

18.
Bioorg Med Chem Lett ; 20(13): 3880-4, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20627562

RESUMEN

MCS-5A, an analog of sangivamycin, selectively inhibits the cyclin-dependent kinases CDK1 and 4 in HL-60 cells in vitro (IC(50): 9.6 and 8.8 1V, respectively), while weakly inhibiting other housekeeping protein kinases. MCS-5A effectively induces HL-60 cell cycle arrest at the G(1) and G(2)/M phases through direct inhibition of CDK1 and 4 activity. In addition, elevated expression of p16(INK4a) and a reduction in the level of hyperphosphorylated pRb showed that 3 1V MCS-5A also induces p16(INK4a)-mediated cell cycle arrest at the G(1) phase. Furthermore, apoptotic induction in MCS-5A-treated HL-60 cells is associated with the release of cytochrome c from mitochondria, which, in turn, results in the activation of procaspase-8, -9 and -3, and the cleavage of poly(ADP-ribose) polymerase (PARP). In addition, the involvement of p16(INK4a) in this apoptotic induction was demonstrated using A549 cells with a homozygous deletion of p16(INK4a). Based on these results, we conclude that MCS-5A is a candidate therapeutic agent for the treatment of human promyelocytic leukemia via the up-regulation of p16(INK4a).


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Citocromos c/metabolismo , Nucleósidos/farmacología , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/antagonistas & inhibidores , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Células HL-60 , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estructura Molecular , Nucleósidos/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Estereoisomerismo , Relación Estructura-Actividad , Regulación hacia Arriba/efectos de los fármacos
19.
Nat Struct Mol Biol ; 12(8): 699-707, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16007099

RESUMEN

Inactivation of the p16(INK4a) tumor suppressor protein is critical for the development of human cancers, including human melanoma. However, the molecular basis of the protein's inhibitory effect on cancer development is not clear. Here we investigated a possible mechanism for p16(INK4a) inhibition of neoplastic transformation and UV-induced skin cancer. We show that p16(INK4a) suppresses the activity of c-Jun N-terminal kinases (JNKs) and that it binds to the glycine-rich loop of the N-terminal domain of JNK3. Although p16(INK4a) does not affect the phosphorylation of JNKs, its interaction with JNK inhibits c-Jun phosphorylation induced by UV exposure. This, in turn, interferes with cell transformation promoted by the H-Ras-JNK-c-Jun-AP-1 signaling axis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Melanoma/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Transducción de Señal/fisiología , Neoplasias Cutáneas/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Glutatión Transferasa , Humanos , Immunoblotting , Ratones , Modelos Moleculares , Fosforilación , Unión Proteica , Técnicas del Sistema de Dos Híbridos
20.
Br J Pharmacol ; 177(10): 2303-2319, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31985814

RESUMEN

BACKGROUND AND PURPOSE: Overexpression or aberrant activation of the T-lymphokine-activated killer cell-originated protein kinase (TOPK) promotes gene expression and growth of solid tumours, implying that TOPK would be a rational target in developing novel anticancer drugs. Acetylshikonin, a diterpenoid compound isolated from Lithospermum erythrorhizon root, exerts a range of biological activities. Here we have investigated whether acetylshikonin, by acting as an inhibitor of TOPK, can attenuate the proliferation of colorectal cancer cells and the growth of patient-derived tumours, in vitro and in vivo. EXPERIMENTAL APPROACH: Targets of acetylshikonin, were identified using kinase profiling analysis, kinetic/binding assay, and computational docking analysis and knock-down techniques. Effects of acetylshikonin on colorectal cancer growth and the underlying mechanisms were evaluated in cell proliferation assays, propidium iodide and annexin-V staining analyses and western blots. Patient-derived tumour xenografts in mice (PDX) and immunohistochemistry were used to assess anti-tumour effects of acetylshikonin. KEY RESULTS: Acetylshikonin directly inhibited TOPK activity, interacting with the ATP-binding pocket of TOPK. Acetylshikonin suppressed cell proliferation by inducing cell cycle arrest at the G1 phase, stimulated apoptosis, and increased the expression of apoptotic biomarkers in colorectal cancer cell lines. Mechanistically, acetylshikonin diminished the phosphorylation and activation of TOPK signalling. Furthermore, acetylshikonin decreased the volume of PDX tumours and reduced the expression of TOPK signalling pathway in xenograft tumours. CONCLUSION AND IMPLICATIONS: Acetylshikonin suppressed growth of colorectal cancer cells by attenuating TOPK signalling. Targeted inhibition of TOPK by acetylshikonin might be a promising new approach to the treatment of colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Células Asesinas Activadas por Linfocinas , Animales , Antraquinonas/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA