Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Drug Metab Dispos ; 52(10): 1094-1103, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39054074

RESUMEN

Equilibrative nucleoside transporters (ENTs) mediate the transmembrane flux of endogenous nucleosides and nucleoside analogs used clinically. The predominant subtype, ENT1, has been well characterized. However, the other subtype, ENT2, has been less well characterized in its native milieu due to its relatively low expression and the confounding influence of coexpressed ENT1. We created a cell model where ENT1 was removed from human embryonic kidney (HEK293) cells using CRISPR/cas9 [ENT1 knockout (KO) cells]; this cell line has ENT2 as the only functional purine transporter. Transporter function was assessed through measurement of [3H]2-chloroadenosine uptake. ENT1 protein was quantified based on the binding of [3H]nitrobenzylthioinosine, and ENT1/ENT2 protein was detected by immunoblotting. Changes in expression of relevant transporters and enzymes involved in purine metabolism were examined by quantitative polymerase chain reaction. Wild-type HEK293 cells and ENT1KO cells had a similar expression of SLC29A2/ENT2 transcript/protein and ENT2-mediated [3H]2-chloroadenosine transport activity (Vmax values of 1.02 ± 0.06 and 1.50 ± 0.22 pmol/µl/s, respectively). Of the endogenous nucleosides/nucleobases tested, adenosine had the highest affinity (Ki) for ENT2 (2.6 µM), while hypoxanthine was the only nucleobase with a submillimolar affinity (320 µM). A range of nucleoside/nucleobase analogs were also tested for their affinity for ENT2 in this model, with affinities (Ki) ranging from 8.6 µM for ticagrelor to 2,300 µM for 6-mercaptopurine. Our data suggest that the removal of endogenous ENT1 from these cells does not change the expression or function of ENT2. This cell line should prove useful for the analysis of novel drugs acting via ENT2 and to study ENT2 regulation. SIGNIFICANCE STATEMENT: We have created a cell line whereby endogenous ENT2 can be studied in detail in the absence of the confounding influence of ENT1. Loss of ENT1 has no impact on the expression and function of ENT2. This novel cell line will provide an ideal model for studying drug interactions with ENT2 as well as the cellular regulation of ENT2 expression and function.


Asunto(s)
Tranportador Equilibrativo 1 de Nucleósido , Transportador Equilibrativo 2 de Nucleósido , Humanos , Células HEK293 , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Tranportador Equilibrativo 1 de Nucleósido/genética , Transportador Equilibrativo 2 de Nucleósido/metabolismo , Transportador Equilibrativo 2 de Nucleósido/genética , Sistemas CRISPR-Cas , 2-Cloroadenosina/farmacología , 2-Cloroadenosina/análogos & derivados , 2-Cloroadenosina/metabolismo , Técnicas de Inactivación de Genes/métodos , Tioinosina/análogos & derivados , Tioinosina/farmacología , Tioinosina/metabolismo , Transporte Biológico/fisiología
2.
J Transl Med ; 22(1): 431, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715059

RESUMEN

BACKGROUND: In humans, two ubiquitously expressed N-myristoyltransferases, NMT1 and NMT2, catalyze myristate transfer to proteins to facilitate membrane targeting and signaling. We investigated the expression of NMTs in numerous cancers and found that NMT2 levels are dysregulated by epigenetic suppression, particularly so in hematologic malignancies. This suggests that pharmacological inhibition of the remaining NMT1 could allow for the selective killing of these cells, sparing normal cells with both NMTs. METHODS AND RESULTS: Transcriptomic analysis of 1200 NMT inhibitor (NMTI)-treated cancer cell lines revealed that NMTI sensitivity relates not only to NMT2 loss or NMT1 dependency, but also correlates with a myristoylation inhibition sensitivity signature comprising 54 genes (MISS-54) enriched in hematologic cancers as well as testis, brain, lung, ovary, and colon cancers. Because non-myristoylated proteins are degraded by a glycine-specific N-degron, differential proteomics revealed the major impact of abrogating NMT1 genetically using CRISPR/Cas9 in cancer cells was surprisingly to reduce mitochondrial respiratory complex I proteins rather than cell signaling proteins, some of which were also reduced, albeit to a lesser extent. Cancer cell treatments with the first-in-class NMTI PCLX-001 (zelenirstat), which is undergoing human phase 1/2a trials in advanced lymphoma and solid tumors, recapitulated these effects. The most downregulated myristoylated mitochondrial protein was NDUFAF4, a complex I assembly factor. Knockout of NDUFAF4 or in vitro cell treatment with zelenirstat resulted in loss of complex I, oxidative phosphorylation and respiration, which impacted metabolomes. CONCLUSIONS: Targeting of both, oxidative phosphorylation and cell signaling partly explains the lethal effects of zelenirstat in select cancer types. While the prognostic value of the sensitivity score MISS-54 remains to be validated in patients, our findings continue to warrant the clinical development of zelenirstat as cancer treatment.


Asunto(s)
Aciltransferasas , Neoplasias , Fosforilación Oxidativa , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/genética , Línea Celular Tumoral , Fosforilación Oxidativa/efectos de los fármacos , Aciltransferasas/metabolismo , Ácido Mirístico/metabolismo , Proteómica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Multiómica
3.
Cancer Rep (Hoboken) ; : e1946, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38064181

RESUMEN

BACKGROUND: Doxorubicin, a first-line anticancer drug for osteosarcoma treatment, has been the subject of recent research exploring the mechanisms behind its chemoresistance and its ability to enhance cell migration at sublethal concentrations. Matrix metalloproteinase-2 (MMP-2), a type IV collagenase and zinc-dependent endopeptidase, is well-known for degrading the extracellular matrix and promoting cancer metastasis. Our previous work demonstrated that nuclear MMP-2 regulates ribosomal RNA transcription via histone clipping, thereby controlling gene expression. Additionally, MMP-2 activity is regulated by the non-receptor tyrosine kinase and oncogene, Src, which plays a crucial role in cell adhesion, invasion, and metastasis. Src kinase is primarily regulated by two endogenous inhibitors: C-terminal Src kinase (Csk) and Csk homologous kinase (CHK/MATK). AIM: In this study, we reveal that the MMP-2 gene acts as an upstream regulator of Src kinase activity by suppressing its endogenous inhibitor, CHK/MATK, in osteosarcoma cells. METHODS AND RESULTS: We show that enhanced osteosarcoma cell migration which is induced by sublethal concentrations of doxorubicin can be overcome by inactivating the MMP-2 gene or overexpressing CHK/MATK. Our findings highlight the MMP-2 gene as a promising additional target for combating cancer cell migration and metastasis. This is due to its role in suppressing on the gene and protein expression of the tumor suppressor CHK/MATK in osteosarcoma. CONCLUSION: By targeting the MMP-2 gene, we can potentially enhance the effectiveness of doxorubicin treatment and reduce chemoresistance in osteosarcoma.

4.
Theranostics ; 12(13): 5675-5690, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35966578

RESUMEN

LPP2 is one of three enzymes in the lipid phosphate phosphatase family (LPP1-3) that dephosphorylate extracellular and intracellular bioactive lipid phosphates and pyrophosphates. LPP2 increases cell growth and LPP2 expression is elevated in a variety of malignancies, implying that LPP2 is a pro-tumorigenic factor. Methods: LPP2 expression in human breast tumors and normal breast tissue was measured by qPCR. To understand the role of LPP2, we knocked out its expression in multiple cell lines using CRISPR/Cas9. Cell proliferation and migration were compared between wild type and LPP2 knockout cells. Cell cycle was measured by flow cytometry, and cell cycle proteins were determined by western blotting. Effects of LPP2 on tumor growth were investigated using syngeneic and xenograft mouse breast cancer models. Results: LPP2 mRNA levels were higher in ER/PR positive, ER/HER2 positive, and triple negative human breast tumors, relative to normal breast tissue. Higher levels of LPP2 in breast tumors, hepatocellular carcinoma, pancreatic adenocarcinoma, and melanomas were prognostic of poorer survival. LPP2 mRNA expression is also increased in Hs-578T, MDA-MB-231, MCF7 and MDA-MB-468 breast cancer cell lines, relative to non-malignant Hs-578Bst, MCF10A and MCF-12A cells. LPP2 knockout in breast cancer cells decreased cell growth by inhibiting G1/S transition, whereas, increasing LPP2 levels in Hs-578Bst and MCF10A cells promoted proliferation. The effects of LPP2 on cell cycle were associated with changes in cyclin A2, cyclin B1, and cell cycle inhibitors, p27 or p21. The level of c-Myc was downregulated by knocking out LPP2, and it was partly restored by re-expressing LPP2. The positive correlation between the expression of LPP2 and c-Myc exists in multiple cancer cell lines including breast, lung, upper aerodigestive tract and urinary tract cancer. LPP2 knockout in MDA-MB-231 or 4T1 cells suppressed tumor formation in mouse breast cancer models, and decreased the in vivo expression of Ki67 and c-Myc of the cancer cells. Conclusion: Targeting LPP2 could provide a new strategy for decreasing c-Myc expression and tumor growth.


Asunto(s)
Adenocarcinoma , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Pancreáticas , Monoéster Fosfórico Hidrolasas/metabolismo , Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Humanos , Ratones , Fosfatidato Fosfatasa , ARN Mensajero
5.
Cell Rep ; 38(11): 110511, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35294884

RESUMEN

An epithelial-to-mesenchymal transition (EMT) phenotype with cancer stem cell-like properties is a critical feature of aggressive/metastatic tumors, but the mechanism(s) that promote it and its relation to metabolic stress remain unknown. Here we show that Collapsin Response Mediator Protein 2A (CRMP2A) is unexpectedly and reversibly induced in cancer cells in response to multiple metabolic stresses, including low glucose and hypoxia, and inhibits EMT/stemness. Loss of CRMP2A, when metabolic stress decreases (e.g., around blood vessels in vivo) or by gene deletion, induces extensive microtubule remodeling, increased glutamine utilization toward pyrimidine synthesis, and an EMT/stemness phenotype with increased migration, chemoresistance, tumor initiation capacity/growth, and metastatic potential. In a cohort of 27 prostate cancer patients with biopsies from primary tumors and distant metastases, CRMP2A expression decreases in the metastatic versus primary tumors. CRMP2A is an endogenous molecular brake on cancer EMT/stemness and its loss increases the aggressiveness and metastatic potential of tumors.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias de la Próstata , Semaforina-3A , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Humanos , Masculino , Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/patología , Semaforina-3A/metabolismo , Estrés Fisiológico
6.
Methods Mol Biol ; 2381: 227-242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34590280

RESUMEN

In addition to advancing the development of gene-editing therapeutics, CRISPR/Cas9 is transforming how functional genetic studies are carried out in the lab. By increasing the ease with which genetic information can be inserted, deleted, or edited in cell and organism models, it facilitates genotype-phenotype analysis. Moreover, CRISPR/Cas9 has revolutionized the speed at which new genes underlying a particular phenotype can be identified through its application in genomic screens. Arrayed high-throughput and pooled lentiviral-based CRISPR/Cas9 screens have now been used in a wide variety of contexts, including the identification of essential genes, genes involved in cancer metastasis and tumor growth, and even genes involved in viral response. This technology has also been successfully used to identify drug targets and drug resistance mechanisms. Here, we provide a detailed protocol for performing a genome-wide pooled lentiviral CRISPR/Cas9 knockout screen to identify genetic modulators of a small-molecule drug. While we exemplify how to identify genes involved in resistance to a cytotoxic histone deacetylase inhibitor, Trichostatin A (TSA), the workflow we present can easily be adapted to different types of selections and other types of exogenous ligands or drugs.


Asunto(s)
Sistemas CRISPR-Cas , Sistemas CRISPR-Cas/genética , Resistencia a Medicamentos , Edición Génica , Genoma , Lentivirus , Preparaciones Farmacéuticas
7.
FEBS J ; 288(23): 6736-6751, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34101354

RESUMEN

Cell proliferation and survival require continuous ribosome biogenesis and protein synthesis. Genes encoding ribosomal RNA are physically located in a specialized substructure within the nucleus known as the nucleolus, which has a central role in the biogenesis of ribosomes. Matrix metalloproteinase-2 was previously detected in the nucleus, however, its role there is elusive. Herein we report that matrix metalloproteinase-2 resides within the nucleolus to regulate ribosomal RNA transcription. Matrix metalloproteinase-2 is enriched at the promoter region of ribosomal RNA gene repeats, and its inhibition downregulates preribosomal RNA transcription. The N-terminal tail of histone H3 is clipped by matrix metalloproteinase-2 in the nucleolus, which is associated with increased ribosomal RNA transcription. Knocking down/out matrix metalloproteinase-2, or inhibiting its activity, prevents histone H3 cleavage and reduces both ribosomal RNA transcription and cell proliferation. In addition to the known extracellular roles of matrix metalloproteinase-2 in tumor growth, our data reveal an epigenetic mechanism whereby intranucleolar matrix metalloproteinase-2 regulates cell proliferation through histone clipping and facilitation of ribosomal RNA transcription.


Asunto(s)
Nucléolo Celular/genética , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Metaloproteinasa 2 de la Matriz/genética , ARN Ribosómico/genética , Transcripción Genética , Línea Celular Tumoral , Nucléolo Celular/metabolismo , Proliferación Celular/genética , Epigénesis Genética , Técnicas de Inactivación de Genes , Humanos , Células MCF-7 , Metaloproteinasa 2 de la Matriz/metabolismo , Microscopía Fluorescente , Células PC-3 , ARN Ribosómico/metabolismo
8.
J Agric Food Chem ; 69(5): 1555-1566, 2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33522796

RESUMEN

Nicotinamide adenine dinucleotide (NAD+) plays a vital role in cellular processes that govern human health and disease. Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in NAD+ biosynthesis. Thus, boosting NAD+ level via an increase in NAMPT levels is an attractive approach for countering the effects of aging and metabolic disease. This study aimed to establish IRW (Ile-Arg-Trp), a small tripeptide derived from ovotransferrin, as a booster of NAMPT levels. Treatment of muscle (L6) cells with IRW increased intracellular NAMPT protein levels (2.2-fold, p < 0.05) and boosted NAD+ (p < 0.01). Both immunoprecipitation and recombinant NAMPT assays indicated the possible NAMPT-activating ability of IRW (p < 0.01). Similarly, IRW increased NAMPT mRNA and protein levels in the liver (2.6-fold, p < 0.01) and muscle tissues (2.3-fold, p < 0.05) of C57BL/6J mice fed with a high-fat diet (HFD). A significantly increased level of circulating NAD+ was also observed following IRW treatment (4.7 fold, p < 0.0001). Dosing of Drosophila melanogaster with IRW elevated both D-NAAM (fly NAMPT) and NAD+ in vivo (p < 0.05). However, IRW treatment did not boost NAMPT levels in SIRT1 KO cells, indicating a possible SIRT1 dependency for the pharmacological effect. Overall, these data indicate that IRW is a novel small peptide booster of the NAMPT pool.


Asunto(s)
Citocinas/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Péptidos/administración & dosificación , Animales , Línea Celular , Citocinas/genética , Drosophila melanogaster , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Obesidad/genética
9.
Molecules ; 25(17)2020 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-32847114

RESUMEN

Resveratrol (3,4,5-Trihydroxy-trans-stilbene) is a naturally occurring polyphenol that exhibits beneficial pleiotropic health effects. It is one of the most promising natural molecules in the prevention and treatment of chronic diseases and autoimmune disorders. One of the key limitations in the clinical use of resveratrol is its extensive metabolic processing to its glucuronides and sulfates. It has been estimated that around 75% of this polyphenol is excreted via feces and urine. To possibly alleviate the extensive metabolic processing and improve bioavailability, we have added segments of acetylsalicylic acid to resveratrol in an attempt to maintain the functional properties of both. We initially characterized resveratrol-aspirin derivatives as products that can inhibit cytochrome P450 Family 1 Subfamily A Member 1 (CYP1A1) activity, DNA methyltransferase (DNMT) activity, and cyclooxygenase (COX) activity. In this study, we provide a detailed analysis of how resveratrol and its aspirin derivatives can inhibit nuclear factor kappa B (NFκB) activation, cytokine production, the growth rate of cancer cells, and in vivo alleviate intestinal inflammation and tumor growth. We identified resveratrol derivatives C3 and C11 as closely preserving resveratrol bioactivities of growth inhibition of cancer cells, inhibition of NFκB activation, activation of sirtuin, and 5' adenosine monophosphate-activated protein kinase (AMPK) activity. We speculate that the aspirin derivatives of resveratrol would be more metabolically stable, resulting in increased efficacy for treating immune disorders and as an anti-cancer agent.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Aspirina , Neoplasias del Colon/tratamiento farmacológico , Inhibidores Enzimáticos , Proteínas de Neoplasias/antagonistas & inhibidores , Resveratrol , Animales , Aspirina/análogos & derivados , Aspirina/química , Aspirina/farmacología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Células HCT116 , Humanos , Ratones , Proteínas de Neoplasias/metabolismo , Resveratrol/análogos & derivados , Resveratrol/química , Resveratrol/farmacología
10.
Biochim Biophys Acta Gen Subj ; 1863(6): 1138-1146, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30980895

RESUMEN

BACKGROUND: Osteoblasts maintain the structural integrity of bone via differentiation and mineralization; therefore, their malfunction or reduced activity can cause serious bone disorders. Although studies have demonstrated the association between nutrients and bone, research on food-derived bioactive peptides and bone health are scanty. METHODS: Osteoblasts MC3T3-E1 were treated with IRW (50 and 25 µM). Cell proliferation, cell cycle, osteoblastic differentiation, and mineralization were tested to evaluate the effects of IRW on osteogenesis promotion. The activation of PI3K-Akt-RUNX2 pathway and collagen synthesis were investigated to better understand the functions of IRW. RESULTS: IRW treatment (50 and 25 µM) in MC3T3-E1 cells caused a significant increase in cell proliferation by increasing the percentage of S and G2/M phase. Furthermore, IRW promoted mineralization in MC3T3-E1 cells. Mechanistically, we found that IRW treatment resulted in a 4-fold increase of Akt serine phosphorylation and a 2-fold increase of its downstream target RUNX2. Expression levels of RUNX2 associated proteins were concomitantly altered: ALP (2-fold increase), Col1A2 (2-fold increase), RANKL (2-fold decrease), and OPG (2-fold increase). Meanwhile, a parallel collagen synthesis pathway was found to contribute to IRW-stimulated osteogenesis. CONCLUSIONS: IRW, an egg-derived small bioactive peptide enhances osteoblastic activity and stimulates osteogenesis. The stimulation is primarily due to the activation of PI3K-Akt-RUNX2 pathway and its downstream effectors, accompanied by a secondary collagen synthesis pathway. GENERAL SIGNIFICANCE: Our results revealed the positive effects of tripeptide IRW on regulating osteogenesis and collagen synthesis, indicating its potential for the prevention or treatment of osteoporosis.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Oligopéptidos/farmacología , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Antígenos de Diferenciación/metabolismo , Línea Celular , Ratones , Osteoblastos/citología
11.
Nat Commun ; 9(1): 1448, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29654299

RESUMEN

Off-target DNA cleavage is a paramount concern when applying CRISPR-Cas9 gene-editing technology to functional genetics and human therapeutic applications. Here, we show that incorporation of next-generation bridged nucleic acids (2',4'-BNANC[N-Me]) as well as locked nucleic acids (LNA) at specific locations in CRISPR-RNAs (crRNAs) broadly reduces off-target DNA cleavage by Cas9 in vitro and in cells by several orders of magnitude. Using single-molecule FRET experiments we show that BNANC incorporation slows Cas9 kinetics and improves specificity by inducing a highly dynamic crRNA-DNA duplex for off-target sequences, which shortens dwell time in the cleavage-competent, "zipped" conformation. In addition to describing a robust technique for improving the precision of CRISPR/Cas9-based gene editing, this study illuminates an application of synthetic nucleic acids.


Asunto(s)
Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN/química , ARN Guía de Kinetoplastida/genética , ARN/química , Línea Celular Tumoral , División del ADN , Endonucleasas/metabolismo , Escherichia coli , Transferencia Resonante de Energía de Fluorescencia , Células HeLa , Humanos , Polimorfismo de Nucleótido Simple , Sensibilidad y Especificidad , Streptococcus pyogenes
12.
J Pharmacol Exp Ther ; 365(2): 354-367, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29555876

RESUMEN

Receptor-interacting protein kinase 2 (RIP2 or RICK, herein referred to as RIPK2) is linked to the pathogen pathway that activates nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) and autophagic activation. Using molecular modeling (docking) and chemoinformatics analyses, we used the RIPK2/ponatinib crystal structure and searched in chemical databases for small molecules exerting binding interactions similar to those exerted by ponatinib. The identified RIPK2 inhibitors potently inhibited the proliferation of cancer cells by > 70% and also inhibited NFκB activity. More importantly, in vivo inhibition of intestinal and lung inflammation rodent models suggests effectiveness to resolve inflammation with low toxicity to the animals. Thus, our identified RIPK2 inhibitor may offer possible therapeutic control of inflammation in diseases such as inflammatory bowel disease, asthma, cystic fibrosis, primary sclerosing cholangitis, and pancreatitis.


Asunto(s)
Descubrimiento de Drogas , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Dominio Catalítico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colitis Ulcerosa/tratamiento farmacológico , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Simulación del Acoplamiento Molecular , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/química , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo
13.
Mol Cancer ; 17(1): 48, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29455673

RESUMEN

The human genome encodes 538 protein kinases that transfer a γ-phosphate group from ATP to serine, threonine, or tyrosine residues. Many of these kinases are associated with human cancer initiation and progression. The recent development of small-molecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy. Significantly, protein kinases are the second most targeted group of drug targets, after the G-protein-coupled receptors. Since the development of the first protein kinase inhibitor, in the early 1980s, 37 kinase inhibitors have received FDA approval for treatment of malignancies such as breast and lung cancer. Furthermore, about 150 kinase-targeted drugs are in clinical phase trials, and many kinase-specific inhibitors are in the preclinical stage of drug development. Nevertheless, many factors confound the clinical efficacy of these molecules. Specific tumor genetics, tumor microenvironment, drug resistance, and pharmacogenomics determine how useful a compound will be in the treatment of a given cancer. This review provides an overview of kinase-targeted drug discovery and development in relation to oncology and highlights the challenges and future potential for kinase-targeted cancer therapies.


Asunto(s)
Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/metabolismo , Animales , Humanos , Estructura Molecular , Neoplasias/tratamiento farmacológico
14.
Science ; 355(6331): 1312-1317, 2017 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-28336669

RESUMEN

DNA repair is essential for life, yet its efficiency declines with age for reasons that are unclear. Numerous proteins possess Nudix homology domains (NHDs) that have no known function. We show that NHDs are NAD+ (oxidized form of nicotinamide adenine dinucleotide) binding domains that regulate protein-protein interactions. The binding of NAD+ to the NHD domain of DBC1 (deleted in breast cancer 1) prevents it from inhibiting PARP1 [poly(adenosine diphosphate-ribose) polymerase], a critical DNA repair protein. As mice age and NAD+ concentrations decline, DBC1 is increasingly bound to PARP1, causing DNA damage to accumulate, a process rapidly reversed by restoring the abundance of NAD+ Thus, NAD+ directly regulates protein-protein interactions, the modulation of which may protect against cancer, radiation, and aging.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Envejecimiento/metabolismo , Reparación del ADN , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Envejecimiento/genética , Animales , Secuencia Conservada , Daño del ADN/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Neoplasias/genética , Neoplasias/metabolismo , Paraquat/farmacología , Poli(ADP-Ribosa) Polimerasa-1/química , Poli(ADP-Ribosa) Polimerasa-1/genética , Dominios y Motivos de Interacción de Proteínas , ARN Interferente Pequeño/genética , Tolerancia a Radiación/genética , Homología de Secuencia de Ácido Nucleico
15.
Cell Rep ; 16(10): 2641-2650, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27568560

RESUMEN

The accumulation of damage caused by oxidative stress has been linked to aging and to the etiology of numerous age-related diseases. The longevity gene, sirtuin 6 (SIRT6), promotes genome stability by facilitating DNA repair, especially under oxidative stress conditions. Here we uncover the mechanism by which SIRT6 is activated by oxidative stress to promote DNA double-strand break (DSB) repair. We show that the stress-activated protein kinase, c-Jun N-terminal kinase (JNK), phosphorylates SIRT6 on serine 10 in response to oxidative stress. This post-translational modification facilitates the mobilization of SIRT6 to DNA damage sites and is required for efficient recruitment of poly (ADP-ribose) polymerase 1 (PARP1) to DNA break sites and for efficient repair of DSBs. Our results demonstrate a post-translational mechanism regulating SIRT6, and they provide the link between oxidative stress signaling and DNA repair pathways that may be critical for hormetic response and longevity assurance.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Sirtuinas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Células HEK293 , Humanos , Ratones Noqueados , Modelos Biológicos , Fosforilación , Fosfoserina/metabolismo
16.
Biochim Biophys Acta ; 1852(6): 1209-18, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25640851

RESUMEN

A number of small molecules with the ability to extend the lifespan of multiple organisms have recently been discovered. Resveratrol, amongst the most prominent of these, has gained widespread attention due to its ability to extend the lifespan of yeast, worms, and flies, and its ability to protect against age-related diseases such as cancer, Alzheimer's, and diabetes in mammals. In this review, we discuss the origins and molecular targets of resveratrol and provide an overview of its effects on the lifespan of simple model organisms and mammals. We also examine the unique ability of resveratrol to extend the healthy years, or healthspan, of mammals and its potential to counteract the symptoms of age-related disease. Finally, we explore the many scientific, medical, and economic challenges faced when translating these findings to the clinic, and examine potential approaches for realizing the possibility of human lifespan extension. This article is part of a Special Issue entitled: Resveratrol: Challenges in translating pre-clinical findings to improved patient outcomes.


Asunto(s)
Longevidad/efectos de los fármacos , Estilbenos/farmacología , Animales , Diseño de Fármacos , Humanos , Resveratrol , Estilbenos/química
17.
Trends Pharmacol Sci ; 35(3): 146-54, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24439680

RESUMEN

Recent studies in mice have identified single molecules that can delay multiple diseases of aging and extend lifespan. In theory, such molecules could prevent dozens of diseases simultaneously, potentially extending healthy years of life. In this review, we discuss recent advances, controversies, opportunities, and challenges surrounding the development of SIRT1 activators, molecules with the potential to delay aging and age-related diseases. Sirtuins comprise a family of NAD⁺-dependent deacylases that are central to the body's response to diet and exercise. New studies indicate that both natural and synthetic sirtuin activating compounds (STACs) work via a common allosteric mechanism to stimulate sirtuin activity, thereby conferring broad health benefits in rodents, primates, and possibly humans. The fact that two-thirds of people in the USA who consume multiple dietary supplements consume resveratrol, a SIRT1 activator, underscores the importance of understanding the biochemical mechanism, physiological effects, and safety of STACs.


Asunto(s)
Envejecimiento/efectos de los fármacos , Sirtuina 1/metabolismo , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Longevidad/efectos de los fármacos , Longevidad/fisiología , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
18.
Cell Cycle ; 12(14): 2233-40, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23892437

RESUMEN

SIRT1 is an NAD (+) -dependent deacetylase that counteracts multiple disease states associated with aging and may underlie some of the health benefits of calorie restriction. Understanding how SIRT1 is regulated in vivo could therefore lead to new strategies to treat age-related diseases. SIRT1 forms a stable complex with DBC1, an endogenous inhibitor. Little is known regarding the biochemical nature of SIRT1-DBC1 complex formation, how it is regulated and whether or not it is possible to block this interaction pharmacologically. In this study, we show that critical residues within the catalytic core of SIRT1 mediate binding to DBC1 via its N-terminal region, and that several carboxamide SIRT1 inhibitors, including EX-527, can completely block this interaction. We identify two acetylation sites on DBC1 that regulate its ability to bind SIRT1 and suppress its activity. Furthermore, we show that DBC1 itself is a substrate for SIRT1. Surprisingly, the effect of EX-527 on SIRT1-DBC1 binding is independent of DBC1 acetylation. Together, these data show that protein acetylation serves as an endogenous regulatory mechanism for SIRT1-DBC1 binding and illuminate a new path to developing small-molecule modulators of SIRT1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carbazoles/farmacología , Regulación de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Procesamiento Proteico-Postraduccional , Sirtuina 1/metabolismo , Acetilación , Proteínas Adaptadoras Transductoras de Señales/genética , Sitios de Unión , Línea Celular Tumoral , Genes Reporteros , Humanos , Luciferasas/genética , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Especificidad por Sustrato
19.
Science ; 339(6124): 1216-9, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23471411

RESUMEN

A molecule that treats multiple age-related diseases would have a major impact on global health and economics. The SIRT1 deacetylase has drawn attention in this regard as a target for drug design. Yet controversy exists around the mechanism of sirtuin-activating compounds (STACs). We found that specific hydrophobic motifs found in SIRT1 substrates such as PGC-1α and FOXO3a facilitate SIRT1 activation by STACs. A single amino acid in SIRT1, Glu(230), located in a structured N-terminal domain, was critical for activation by all previously reported STAC scaffolds and a new class of chemically distinct activators. In primary cells reconstituted with activation-defective SIRT1, the metabolic effects of STACs were blocked. Thus, SIRT1 can be directly activated through an allosteric mechanism common to chemically diverse STACs.


Asunto(s)
Sirtuina 1/química , Sirtuina 1/metabolismo , Estilbenos/farmacología , Regulación Alostérica , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Células Cultivadas , Activación Enzimática , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Ácido Glutámico/química , Ácido Glutámico/genética , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Datos de Secuencia Molecular , Mioblastos/efectos de los fármacos , Mioblastos/enzimología , Estructura Terciaria de Proteína , Resveratrol , Sirtuina 1/genética , Estilbenos/química , Especificidad por Sustrato
20.
Cell Cycle ; 12(2): 263-70, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23255128

RESUMEN

SIRT1 is an evolutionarily conserved protein deacetylase that modulates stress response, cellular metabolism and aging in model organisms. While SIRT1 exerts beneficial effects in protecting against age-related diseases, the role of SIRT1 in cancer has been controversial. SIRT1 promotes cell survival by deacetylating, and thereby negatively regulating the activity of important tumor suppressors such as p53. In this regard, SIRT1 has been considered to be a potential oncogene, and SIRT1 inhibitors have been studied for possible anticancer therapeutic effects. In contrast, it has been shown that SIRT1 deficiency leads to increased genomic instability and tumorigenesis, and that overexpression of SIRT1 attenuates cancer formation in mice, suggesting it may also act as a tumor suppressor. Based on this evidence, SIRT1-activating molecules could act as candidate chemotherapeutic drugs. In order to gain insight into the role of SIRT1 in cancer, we performed a comprehensive resequencing analysis of the SIRT1 gene in 41 tumor cell lines and found an unusually excessive homozygosity, which was confirmed to be allelic loss by microsatellite analysis. Furthermore, we found two novel SIRT1 mutations (D739Y and R65_A72del) in addition to the known, rare non-synonymous variation resulting in I731V. In vitro assays using purified SIRT1 protein showed that these mutations do not alter SIRT1 deacetylase activity or telomerase activity, which was shown to be regulated by SIRT1. We conclude that allelic loss or mutations in the SIRT1 gene occur prevalently during tumorigenesis, supporting the assertion that SIRT1 may serve as a tumor suppressor.


Asunto(s)
Transformación Celular Neoplásica/genética , Pérdida de Heterocigocidad/genética , Mutación/genética , Sirtuina 1/genética , Western Blotting , Línea Celular Tumoral , Análisis Mutacional de ADN , Cartilla de ADN/genética , Humanos , Repeticiones de Microsatélite/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA