Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 220
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Semin Cancer Biol ; 83: 335-352, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-33453404

RESUMEN

In-depth insights in cancer biology over the past decades have highlighted the important roles of epigenetic mechanisms in the initiation and progression of tumorigenesis. The cancer epigenome usually experiences multiple alternations, including genome-wide DNA hypomethylation and site-specific DNA hypermethylation, various histone posttranslational modifications, and dysregulation of non-coding RNAs (ncRNAs). These epigenetic changes are plastic and reversible, and could potentially occur in the early stage of carcinogenesis preceding genetic mutation, offering unique opportunities for intervention therapies. Therefore, targeting the cancer epigenome or cancer epigenetic dysregulation with some selected agents (called epi-drugs) represents an evolving and promising strategy for cancer chemoprevention and therapy. Phytochemicals, as a class of pleiotropic molecules, have manifested great potential in modulating different cancer processes through epigenetic machinery, of which green tea polyphenol epigallocatechin-3-gallate (EGCG) is one of the most extensively studied. In this review, we first summarize epigenetic events involved in the pathogenesis of cancer, including DNA/RNA methylations, histone modifications and ncRNAs' dysregulations. We then focus on the recently discovered roles of phytochemicals, with a special emphasis on EGCG, in modulating different cancer processes through regulating epigenetic machinery. We finally discuss limitations of EGCG as an epigenetic modulator for cancer chemoprevention and treatment and offer potential strategies to overcome the shortcomings.


Asunto(s)
Neoplasias , , Catequina/análogos & derivados , Epigénesis Genética , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Polifenoles/farmacología , Polifenoles/uso terapéutico , Té/química
2.
Nature ; 552(7684): 194-199, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29211715

RESUMEN

Cancer incidence is rising and this global challenge is further exacerbated by tumour resistance to available medicines. A promising approach to meet the need for improved cancer treatment is drug repurposing. Here we highlight the potential for repurposing disulfiram (also known by the trade name Antabuse), an old alcohol-aversion drug that has been shown to be effective against diverse cancer types in preclinical studies. Our nationwide epidemiological study reveals that patients who continuously used disulfiram have a lower risk of death from cancer compared to those who stopped using the drug at their diagnosis. Moreover, we identify the ditiocarb-copper complex as the metabolite of disulfiram that is responsible for its anti-cancer effects, and provide methods to detect preferential accumulation of the complex in tumours and candidate biomarkers to analyse its effect on cells and tissues. Finally, our functional and biophysical analyses reveal the molecular target of disulfiram's tumour-suppressing effects as NPL4, an adaptor of p97 (also known as VCP) segregase, which is essential for the turnover of proteins involved in multiple regulatory and stress-response pathways in cells.


Asunto(s)
Disuasivos de Alcohol , Alcoholismo/tratamiento farmacológico , Antineoplásicos , Disulfiram/farmacología , Disulfiram/uso terapéutico , Reposicionamiento de Medicamentos , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Adulto , Disuasivos de Alcohol/farmacología , Disuasivos de Alcohol/uso terapéutico , Alcoholismo/epidemiología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Cobre/química , Dinamarca/epidemiología , Disulfiram/química , Femenino , Respuesta al Choque Térmico/efectos de los fármacos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Terapia Molecular Dirigida , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Proteínas Nucleares/química , Agregado de Proteínas , Unión Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos
3.
Int J Mol Sci ; 24(1)2023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-36614319

RESUMEN

Activated Leukocyte Cell Adhesion Molecule (ALCAM/CD166) is a cell-cell adhesion protein conferring heterotypic and homotypic interactions between cells of the same type and different types. It is aberrantly expressed in various cancer types and has been shown to be a regulator of cancer metastasis. In the present study, we investigated potential roles of ALCAM in the peritoneal transcoelomic metastasis in gastrointestinal cancers, a metastatic type commonly occurred in gastro-intestinal and gynaecological malignancies and resulting in poor clinical outcomes. Specifically, we studied whether ALCAM acts as both a 'seed' receptor in these tumour cells and a 'soil' receptor in peritoneal mesothelial cells during cancer metastasis. Gastric cancer and pancreatic cancer tissues with or without peritoneal metastasis were compared for their levels of ALCAM expression. The impact of ALCAM expression in these tumours was also correlated to the patients' clinical outcomes, namely peritoneal metastasis-free survival. In addition, cancer cells of gastric and pancreatic origins were used to create cell models with decreased or increased levels of ALCAM expression by genetic knocking down or overexpression, respectively. Human peritoneal mesothelial cells were also genetically transfected to generate cell models with different profiles of ALCAM expression. These cell models were used in the tumour-mesothelial interaction assay to assess if and how the interaction was influenced by ALCAM. Both gastric and pancreatic tumour tissues from patients who developed peritoneal metastases had higher levels of ALCAM transcript than those without. Patients who had tumours with high levels of ALCAM had a much shorter peritoneal metastasis free survival compared with those who had low ALCAM expression (p = 0.006). ALCAM knockdown of the mesothelial cell line MET5A rendered the cells with reduced interaction with both gastric cancer cells and pancreatic cancer cells. Likewise, levels of ALCAM in both human gastric and pancreatic cancer cells were also a determining factor for their adhesiveness to mesothelial cells, a process that was likely to be triggered the phosphorylation of the SRC kinase. A soluble ALCAM (sALCAM) was found to be able to inhibit the adhesiveness between cancer cells and mesothelial cells, mechanistically behaving like a SRC kinase inhibitor. ALCAM is an indicator of peritoneal metastasis in both gastric and pancreatic cancer patients. It acts as not only a potential peritoneal 'soil' receptor of tumour seeding but also a 'soil' receptor in peritoneal mesothelial cells during cancer metastasis. These findings have an important therapeutic implication for treating peritoneal transcoelomic metastases.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado , Neoplasias Pancreáticas , Neoplasias Peritoneales , Neoplasias Gástricas , Humanos , Molécula de Adhesión Celular del Leucocito Activado/genética , Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Adhesión Celular , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Familia-src Quinasas/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Peritoneales/secundario , Neoplasias Pancreáticas
4.
Chin J Cancer Res ; 35(4): 365-385, 2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37691891

RESUMEN

Objective: Striatins (STRNs) family, which contains three multi-domain scaffolding proteins, are cornerstones of the striatins interacting phosphatase and kinase (STRIPAK) complex. Although the role of the STRIPAK complex in cancer has become recognized in recent years, its clinical significance in breast cancer has not been fully established. Methods: Using a freshly frozen breast cancer tissue cohort containing both cancerous and adjacent normal mammary tissues, we quantitatively evaluated the transcript-level expression of all members within the STRIPAK complex along with some key interacting and regulatory proteins of STRNs. The expression profile of each molecule and the integrated pattern of the complex members were assessed against the clinical-pathological factors of the patients. The Cancer Genome Atlas (TCGA) dataset was used to evaluate the breast cancer patients' response to chemotherapies. Four human breast cancer cell lines, MDA-MB-231, MDA-MB-361, MCF-7, and SK-BR-3, were subsequently adopted for in vitro work. Results: Here we found that high-level expressions of STRIP2, calmodulin, CCM3, MINK1 and SLMAP were respectively associated with shorter overall survival (OS) of patients. Although the similar pattern observed for STRN3, STRN4 and a contrary pattern observed for PPP2CA, PPP2CB and PPPR1A were not significant, the integrated expression profile of STRNs group and PPP2 group members constitutes a highly significant prognostic indicator for OS [P<0.001, hazard ratio (HR)=2.04, 95% confidence interval (95% CI), 1.36-3.07] and disease-free survival (DFS) (P=0.003, HR=1.40, 95% CI, 1.12-1.75). Reduced expression of STRN3 has an influence on the biological functions including adhesiveness and migration. In line with our clinical findings, the breast cancer cells responded to STRN3 knockdown with changes in their chemo-sensitivity, of which the response is also breast cancer subtype dependent. Conclusions: Our results suggest a possible role of the STRIPAK complex in breast cancer development and prognosis. Among the members, the expression profile of STRN3 presents a valuable factor for assessing patients' responses to drug treatment.

5.
Semin Cancer Biol ; 68: 105-122, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31883910

RESUMEN

The ubiquitin-proteasome system (UPS) plays a central role in the degradation of cellular proteins. Targeting protein degradation has been validated as an effective strategy for cancer therapy since 2003. Several components of the UPS have been validated as potential anticancer targets, including 20S proteasomes, 19S proteasome-associated deubiquitinases (DUBs) and ubiquitin ligases (E3s). 20S proteasome inhibitors (such as bortezomib/BTZ and carfilzomib/CFZ) have been approved by the U.S. Food and Drug Administration (FDA) for the treatment of multiple myeloma (MM) and some other liquid tumors. Although survival of MM patients has been improved by the introduction of BTZ-based therapies, these clinical 20S proteasome inhibitors have several limitations, including emergence of resistance in MM patients, neuro-toxicities, and little efficacy in solid tumors. One of strategies to improve the current status of cancer treatment is to repurpose old drugs with UPS-inhibitory properties as new anticancer agents. Old drug reposition represents an attractive drug discovery approach compared to the traditional de novo drug discovery process which is time-consuming and costly. In this review, we summarize status of repurposed inhibitors of various UPS components, including 20S proteasomes, 19S-associated DUBs, and ubiquitin ligase E3s. The original and new mechanisms of action, molecular targets, and potential anticancer activities of these repurposed UPS inhibitors are reviewed, and their new uses including combinational therapies for cancer treatment are discussed.


Asunto(s)
Antineoplásicos/uso terapéutico , Descubrimiento de Drogas , Reposicionamiento de Medicamentos/métodos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/uso terapéutico , Animales , Humanos , Ubiquitina/metabolismo
6.
Bioorg Med Chem ; 30: 115931, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33341501

RESUMEN

The ubiquitin-proteasome system (UPS) plays an important role in maintaining protein homeostasis by degrading intracellular proteins. In the proteasome, poly-ubiquitinated proteins are deubiquitinated by three deubiquitinases (DUBs) associated with 19S regulatory particle before degradation via 20S core particle. Ubiquitin carboxyl-terminal hydrolase L5 (UCHL5) is one of three proteasome-associated DUBs that control the fate of ubiquitinated substrates implicated in cancer survival and progression. In this study, we have performed virtual screening of an FDA approved drug library with UCHL5 and discovered tiaprofenic acid (TA) as a potential binder. With molecular docking analysis and in-vitro DUB assay, we have designed, synthesized, and evaluated a series of TA derivatives for inhibition of UCHL5 activity. We demonstrate that one TA derivative, TAB2, acts as an inhibitor of UCHL5.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Propionatos/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Propionatos/síntesis química , Propionatos/química , Relación Estructura-Actividad , Ubiquitina Tiolesterasa/metabolismo
7.
J Cell Biochem ; 120(8): 14065-14075, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30963630

RESUMEN

The levels of organic pollutants, such as optical brightener (OB) compounds, in the global environment have been increasing in recent years. The toxicological effects and signal transduction systems associated with OB toxicity have not been thoroughly studied. The ubiquitin-proteasome system (UPS) plays a crucial role in regulating multiple essential cellular processes, and proteasome-associated cysteine deubiquitinases (DUBs), ubiquitin C-terminal hydrolase L5 (UCHL5) and USP14, are two major regulators for (de)ubiquitination and stability of many important target proteins. Therefore, potential inhibition of UCHL5 and USP14 activities by some environmental chemicals might cause in vivo toxicity. In the current study we hypothesize that electrophilic OB compounds, such as 4,4'-diamino-2,2'-stilbenedisulfonic acid(DAST), fluorescent brightener 28 (FB-28) and FB-71, can interact with the catalytic triads (CYS, HIS, and ASP) of UCHL5 and USP14 and inhibit their enzymatic activities, leading to cell growth suppression. This hypothesis is supported by our findings presented in this study. Results from in silico computational docking and ubiquitin vinyl sulfone assay confirmed the UCHL5/USP14-inhibitory activities of these OB compounds that have potencies in an order of: FB-71 > FB-28 > DAST. Furthermore, inhibition of these two proteasomal DUBs by OBs resulted in cell growth inhibition and apoptosis induction in two human breast cancer cell models. In addition, we found that OB-mediated DUB inhibition triggers a feedback reaction in which expression of UCHL5 and USP14 proteins is increased to compromise the suppressed activities. Our study suggests that these commonly used OB compounds may target and inhibit proteasomal cysteine DUBs, which should contribute to their toxicological effects in vivo.


Asunto(s)
Cisteína/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Contaminantes Ambientales/toxicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Apoptosis/efectos de los fármacos , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Contaminantes Ambientales/química , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo
8.
J Cell Biochem ; 119(11): 9006-9016, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30015387

RESUMEN

Isothiocyanates (ITCs) are natural chemoprotective products found abundantly in cruciferous vegetables. However, the cancer-relevant targets and molecular mechanisms of ITCs remain unclear. We hypothesize that ITCs, as electrophiles, can interact with the catalytic triads (CYS, HIS, and ASP) of the proteasomal cysteine deubiquitinases USP14 and UCHL5, ultimately inhibiting their activities. In the current study, we exploited this possibility by performing both computational docking and biochemical validation assays using human breast and prostate cancer cell models. Docking results suggest that benzyl isothiocyanate, phenethyl isothiocyanate, and DL-sulforaphane are more potent inhibitors of UCHL5 than USP14, and these ITCs could interact with the catalytic triads of UCHL5 and USP14. Indeed, ubiquitin vinyl sulfone assay confirmed the inhibitory activity of each ITC on the ubiquitin-binding activity of UCHL5 and USP14. We also found that inhibition of USP-14 and UCHL5 activities by the ITCs caused increased levels of USP14 and UCHL5 proteins, but not the third 19S-deubiquitinating enzyme (DUB), POH1/RPN11, suggesting feedback loop activation and further supporting that ITCs are inhibitors of proteasomal cysteine DUBs. Associated with DUB inhibition by ITCs, ubiquitinated proteins were significantly increased, accompanied with induction of apoptosis, inhibition of proliferation and suppression of cell invasion. Our findings of ITCs as proteasomal cysteine DUB inhibitors should provide insightful information for designing, discovering and developing potent, specific 19S-DUB inhibitors for cancer therapies.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Isotiocianatos/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Biología Computacional , Humanos , Masculino , Transactivadores/metabolismo , Ubiquitina Tiolesterasa/metabolismo
9.
Cancer Metastasis Rev ; 36(4): 717-736, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29047025

RESUMEN

In the past 15 years, the proteasome has been validated as an anti-cancer drug target and 20S proteasome inhibitors (such as bortezomib and carfilzomib) have been approved by the FDA for the treatment of multiple myeloma and some other liquid tumors. However, there are shortcomings of clinical proteasome inhibitors, including severe toxicity, drug resistance, and no effect in solid tumors. At the same time, extensive research has been conducted in the areas of natural compounds and old drug repositioning towards the goal of discovering effective, economical, low toxicity proteasome-inhibitory anti-cancer drugs. A variety of dietary polyphenols, medicinal molecules, metallic complexes, and metal-binding compounds have been found to be able to selectively inhibit tumor cellular proteasomes and induce apoptotic cell death in vitro and in vivo, supporting the clinical success of specific 20S proteasome inhibitors bortezomib and carfilzomib. Therefore, the discovery of natural proteasome inhibitors and researching old drugs with proteasome-inhibitory properties may provide an alternative strategy for improving the current status of cancer treatment and even prevention.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Ubiquitina/antagonistas & inhibidores , Animales , Reposicionamiento de Medicamentos , Humanos , Terapia Molecular Dirigida , Neoplasias/enzimología , Ubiquitina/metabolismo
10.
Cancer Metastasis Rev ; 36(4): 655-668, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29039082

RESUMEN

Deubiquitinases (DUBs) play an important role in protein quality control in eukaryotic cells due to their ability to specifically remove ubiquitin from substrate proteins. Therefore, recent findings have focused on the relevance of DUBs to cancer development, and pharmacological intervention on these enzymes has become a promising strategy for cancer therapy. In particular, several DUBs are physically and/or functionally associated with the proteasome and are attractive targets for the development of novel anticancer drugs. The successful clinical application of cisplatin in cancer treatment has prompted researchers to develop various metal-based anticancer agents with new properties. Recently, we have reported that several metal-based drugs, such as the antirheumatic gold agent auranofin (AF), the antifouling paint biocides copper pyrithione (CuPT) and zinc pyrithione (ZnPT), and also our two synthesized complexes platinum pyrithione (PtPT) and nickel pyrithione (NiPT), can target the proteasomal DUBs UCHL5 and USP14. In this review, we summarize the recently reported small molecule inhibitors of proteasomal DUBs, with a focus on discussion of the unique nature of metal-based proteasomal DUB inhibitors and their anticancer activity.


Asunto(s)
Antineoplásicos/farmacología , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Compuestos Organometálicos/farmacología , Inhibidores de Proteasoma/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Enzimas Desubicuitinizantes/metabolismo , Humanos
11.
J Cell Biochem ; 118(9): 2722-2730, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28165150

RESUMEN

Pancreatic cancer is the fourth leading cause of cancer-related death in the United States. While Gemcitabine-based chemotherapy is the first-line treatment for locally advanced pancreatic cancer, its resistance is a large obstacle in the field. Understanding the molecular basis of gemcitabine resistance is therefore critical for increasing the efficacy of gemcitabine-based chemotherapy and improving the survival rate of the cancer patients. Here, we investigated the sensitivity of a pair of established human pancreatic cancer cell lines, MIAPaCa-E (relatively sensitive to gemcitabine, called GS), and MIAPaCa-M (highly resistant to gemcitabine, called GR) to various inhibitors of EGFR or PI3K/AKT/mTOR pathways. We found that compared to GS cells, GR cells were resistant to all the tested inhibitors of EGFR, AKT, and PI3K, but were equally, if not more sensitive to the mTOR inhibitor everolimus. Mechanistically, everolimus treatment decreased levels of total mTOR and Ser2248-phosphorylated mTOR protein in GR over GS cells, suggesting greater inhibition of mTOR by everolimus in GR cells. High levels of Thr389- and Thr371-phosphorylated p70S6 protein (a mTOR substrate) were found in GR cells treated with gemcitabine, but not everolimus, supporting that inhibition of p70S6 protein phosphorylation by everolimus is associated with its ability to overcome gemcitabine resistance. Everolimus also induced higher levels of caspase-3/-7 activation in GR over GS cells, and everolimus-mediated mTOR inhibition lead to G2 arrest in GR cells but G1 arrest in GS cells. Our study suggests the a mTOR inhibitor could improve the efficacy of gemcitabine-based chemotherapy for otherwise resistant pancreatic cancer. J. Cell. Biochem. 118: 2722-2730, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Everolimus/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Gemcitabina
12.
J Cell Biochem ; 118(5): 1239-1248, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27813130

RESUMEN

Although effective for the treatment of hematological malignancies, the FDA approved proteasome inhibitors bortezomib and carfilzomib have limited efficacy in solid tumors including triple negative breast cancer (TNBC). Chemotherapy is the only option for treating TNBC due to the absence of specific therapeutic targets. Therefore, development of new TNBC therapeutic strategies has been warranted. We studied whether P-glycoprotein (P-gp) inhibition could sensitize TNBC cells to proteasome inhibitors. When verapamil, a P-gp inhibitor, was combined with the proteasome inhibitor MG132, bortezomib, or carfilzomib, the cytotoxic effects and apoptosis in TNBC MDA-MB-231 cells were enhanced, compared to each treatment alone. Furthermore, addition of verapamil improved proteasome-inhibitory properties of MG132, bortezomib, or carfilzomib in MDA-MB-231 cells, as shown by the increased accumulation of ubiquitinated proteins and proteasome substrates such as IκBα and p27kip1 . Additionally, when nicardipine, another P-gp inhibitor, was combined with bortezomib or carfilzomib, enhanced inhibition of MDA-MB-231 cell proliferation was observed. These findings indicate that P-gp inhibitors could sensitize TNBC cells to structurally and functionally diverse proteasome inhibitors and might provide new treatment strategy for TNBC. J. Cell. Biochem. 118: 1239-1248, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Antineoplásicos/farmacología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Inhibidores de Proteasoma/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Verapamilo/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ubiquitinación
13.
Tetrahedron Lett ; 58(19): 1895-1899, 2017 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-31798193

RESUMEN

Five bioactive Annonaceous acetogenins, including three new compounds, annonamuricins A (1), B (2), and C (3), one registered but no spectral data reported compounds, annonamuricin D (4), and one known compound annonacin (5) were isolated from Graviola fruit (Annona muricata) and further determined through bioassay-guided fractionation. All five compounds are C35 Anonnonaceous acetogenins with a mono-tetrahydrofuran ring and four hydroxyls. Their structures were elucidated using spectral methods as well as chemical modification after isolation via chromatographic techniques and HPLC purification. These acetogenins demonstrated potent anti-proliferative activities against human prostate cancer PC-3 cells.

14.
Semin Cancer Biol ; 35 Suppl: S78-S103, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25936818

RESUMEN

Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.


Asunto(s)
Apoptosis/genética , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/genética , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
15.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26590477

RESUMEN

Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.


Asunto(s)
Heterogeneidad Genética , Terapia Molecular Dirigida , Neoplasias/terapia , Medicina de Precisión , Antineoplásicos Fitogénicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias/genética , Neoplasias/patología , Neoplasias/prevención & control , Transducción de Señal , Microambiente Tumoral/genética
16.
J Cell Biochem ; 117(1): 144-51, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26084403

RESUMEN

The ubiquitin-proteasome pathway has gained attention as a potential chemotherapeutic target, owing to its importance in the maintenance of protein homeostasis and the observation that cancer cells are more dependent on this pathway than normal cells. Additionally, inhibition of histone deacetylases (HDACs) by their inhibitors like Vorinostat (SAHA) has also proven a useful strategy in cancer therapy and the concomitant use of proteasome and HDAC inhibitors has been shown to be superior to either treatment alone. It has also been reported that delta-aminolevulinic acid dehydratase (ALAD) is a proteasome-associated protein, and may function as an endogenous proteasome inhibitor. While the role of ALAD in the heme biosynthetic pathway is well characterized, little is known about its interaction with, and the mechanism by which it inhibits, the proteasome. In the present study, this ALAD-proteasome complex was further characterized in cultured prostate cancer cells and the effects of SAHA treatment on the regulation of ALAD were investigated. ALAD interacts with the 20S proteasomal core, but not the 19S regulatory cap. Some ubiquitinated species were detected in ALAD immunoprecipitates that have similar molecular weights to ubiquitinated proteasomal α2 subunits, suggesting preferred binding of ALAD to ubiquitinated α2. Additionally, SAHA treatment increases levels of ALAD protein and an acetylated protein with a molecular weight similar to the ubiquitinated α2 subunit. Thus, the results of this study suggest that ALAD may play a regulatory role in a previously unreported post-translational modification of proteasomal α subunits.


Asunto(s)
Porfobilinógeno Sintasa/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Acetilación/efectos de los fármacos , Animales , Bovinos , Línea Celular Tumoral , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ubiquitinación/efectos de los fármacos , Vorinostat
17.
J Cell Biochem ; 117(10): 2357-69, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26950525

RESUMEN

Uterine fibroids (leiomyomas) are very common benign tumors grown on the smooth muscle layer of the uterus, present in up to 75% of reproductive-age women and causing significant morbidity in a subset of this population. Although the etiology and biology of uterine fibroids are unclear, strong evidence supports that cell proliferation, angiogenesis and fibrosis are involved in their formation and growth. Currently the only cure for uterine fibroids is hysterectomy; the available alternative therapies have limitations. Thus, there is an urgent need for developing a novel strategy for treating this condition. The green tea polyphenol epigallocatechin gallate (EGCG) inhibits the growth of uterine leiomyoma cells in vitro and in vivo, and the use of a green tea extract (containing 45% EGCG) has demonstrated clinical activity without side effects in women with symptomatic uterine fibroids. However, EGCG has a number of shortcomings, including low stability, poor bioavailability, and high metabolic transformations under physiological conditions, presenting challenges for its development as a therapeutic agent. We developed a prodrug of EGCG (Pro-EGCG or 1) which shows increased stability, bioavailability and biological activity in vivo as compared to EGCG. We also synthesized prodrugs of EGCG analogs, compounds 2a and 4a, in order to potentially reduce their susceptibility to methylation/inhibition by catechol-O-methyltransferase. Here, we determined the effect of EGCG, Pro-EGCG, and 2a and 4a on cultured human uterine leiomyoma cells, and found that 2a and 4a have potent antiproliferative, antiangiogenic, and antifibrotic activities. J. Cell. Biochem. 117: 2357-2369, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Apoptosis/efectos de los fármacos , Catequina/análogos & derivados , Leiomioma/patología , Neovascularización Patológica/patología , Profármacos/farmacología , Té/química , Neoplasias Uterinas/patología , Western Blotting , Catequina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Leiomioma/tratamiento farmacológico , Leiomioma/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/metabolismo
18.
Bioorg Med Chem Lett ; 26(17): 4382-5, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27499453

RESUMEN

Bioassay-guided fractionation of the fruit powder of Graviola (Annona muricata) was continued to be conducted and yielded three more novel bioactive compounds: C-35 annonaceous acetogenins, muricins M and N, and C-37 annonaceous acetogenins, muricenin. They all contain a mono-tetrahydrofuran ring and four hydroxyl groups. The structures were elucidated by spectral methods and chemical modification after isolation via open column chromatographic separation and HPLC purification. Especially, murices M and N demonstrated more potent anti-proliferative activities against human prostate cancer PC-3 cells.


Asunto(s)
Acetogeninas/química , Acetogeninas/farmacología , Annona/química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Estructura Molecular , Neoplasias de la Próstata/tratamiento farmacológico
19.
Breast Cancer Res Treat ; 153(1): 79-88, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26227473

RESUMEN

The purpose of present study is to examine the mechanism of the 5'-AMP-activated protein kinase (AMPK) activation induced by proteasome inhibitors. AMPK activation and ubiquitin proteasome system (UPS) inhibition have gained great attention as therapeutic strategies for the treatment of certain types of cancers. While AMPK serves as a master regulator of cellular metabolism, UPS regulates protein homeostasis. However, the relationship between these two important pathways is not very clear. We observe that proteasome inhibition leads to AMPK activation in human breast cancer cells. siRNA transfection, western blotting, qPCR, and proteasomal inhibition assays were used to study the mechanism of proteasome inhibitor-induced AMPK activation using human triple-negative breast cancer, lung, and cervical cancer cell lines. We report that a variety of proteasome inhibitors activate AMPK in all the tested different cancer cell lines. Our data using liver kinase B1-deficient cancer cells suggest that proteasome inhibitor-induced AMPK activation is primarily mediated by Calcium/Calmodulin-dependent kinase kinase ß (CaMKKß). This hypothesis is supported by that pharmacological or genetic inhibition of CaMKKß leads to a decrease in proteasome inhibitor-induced AMPK activation. Additionally, the AMPK-activating function of the FDA-approved proteasome inhibitor bortezomib depends on an increase in intracellular calcium levels as calcium chelation abrogates its induced AMPK activation. Finally, bortezomib-mediated upregulation in CaMKKß levels is due to its enhanced protein synthesis. These data suggest that proteasome inhibitors indirectly activate AMPK in human cancer cells primarily via Ca(2+)-CaMKKß-dependent pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Inhibidores de Proteasoma/farmacología , Quinasas de la Proteína-Quinasa Activada por el AMP , Bortezomib/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Línea Celular Tumoral , Quimotripsina , Activación Enzimática/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos
20.
J Biol Inorg Chem ; 20(1): 109-121, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25467055

RESUMEN

Ubiquitin-proteasome system (UPS) plays a crucial role in many cellular processes such as cell cycle, proliferation and apoptosis. Aberrant activation of UPS may result in cellular transformation or other altered pathological conditions. Previous studies have shown that metal-based complexes could inhibit proteasome activity and induce apoptosis in certain human cancer cells. In the current study, we report that the cadmium and copper complexes with heterocycle-ornithine Schiff base are potent inhibitors of proteasomal chymotrypsin-like (CT-like) activity, leading to induction of apoptosis in cancer cells. Two novel copper-containing complexes and two novel cadmium-containing complexes with different heterocycle-ornithine Schiff base structures as ligands were synthesized and characterized. We found that complexes Cu1, Cd1 and Cd2 show proteasome-inhibitory activities in human breast cancer MDA-MB-231 and human prostate cancer LNCaP cells, resulting in the accumulation of p27, a natural proteasome substrate and other ubiquitinated proteins, followed by the induction of apoptosis. Our results suggest that metal complexes with heterocycle-ornithine Schiff base have proteasome-inhibitory capabilities and have the potential to be developed into novel anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Complejos de Coordinación/farmacología , Inhibidores de Proteasoma/farmacología , Antineoplásicos/química , Cadmio/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/química , Cobre/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Ornitina/química , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/química , Bases de Schiff
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA