Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Biol Chem ; 294(3): 827-837, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30487288

RESUMEN

Type I interferons (IFNs) induce expression of multiple genes that control innate immune responses to invoke both antiviral and antineoplastic activities. Transcription of these interferon-stimulated genes (ISGs) occurs upon activation of the canonical Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathways. Phosphorylation and acetylation are both events crucial to tightly regulate expression of ISGs. Here, using mouse embryonic fibroblasts and an array of biochemical methods including immunoblotting and kinase assays, we show that sirtuin 2 (SIRT2), a member of the NAD-dependent protein deacetylase family, is involved in type I IFN signaling. We found that SIRT2 deacetylates cyclin-dependent kinase 9 (CDK9) in a type I IFN-dependent manner and that the CDK9 deacetylation is essential for STAT1 phosphorylation at Ser-727. We also found that SIRT2 is subsequently required for the transcription of ISGs and for IFN-driven antiproliferative responses in both normal and malignant cells. These findings establish the existence of a previously unreported signaling pathway whose function is essential for the control of JAK-STAT signaling and the regulation of IFN responses. Our findings suggest that targeting sirtuin activities may offer an avenue in the development of therapies for managing immune-related diseases and cancer.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Interferón Tipo I/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Sirtuina 2/metabolismo , Acetilación , Animales , Quinasa 9 Dependiente de la Ciclina/genética , Humanos , Interferón Tipo I/genética , Ratones , Ratones Noqueados , Fosforilación , Factor de Transcripción STAT1/genética , Sirtuina 2/genética , Transcripción Genética , Células U937
2.
Mol Cell ; 32(1): 11-20, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18851829

RESUMEN

Germline mutations of BRCA1 predispose women to breast and ovarian cancers. However, the downstream mediators of BRCA1 function in tumor suppression remain elusive. We found that human BRCA1-associated breast cancers have lower levels of SIRT1 than their normal controls. We further demonstrated that mammary tumors from Brca1 mutant mice have low levels of Sirt1 and high levels of Survivin, which is reversed by induced expression of Brca1. BRCA1 binds to the SIRT1 promoter and increases SIRT1 expression, which in turn inhibits Survivin by changing the epigenetic modification of histone H3. Absence of SIRT1 blocks the regulation of Survivin by BRCA1. Furthermore, we demonstrated that activation of Sirt1 and inhibition of Survivin expression by resveratrol elicit a more profound inhibitory effect on Brca1 mutant cancer cells than on Brca1-wild-type cancer cells both in vitro and in vivo. These findings suggest that resveratrol treatment serves as an excellent strategy for targeted therapy for BRCA1-associated breast cancer.


Asunto(s)
Genes BRCA1 , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Proteína BRCA1/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Epigénesis Genética , Femenino , Mutación de Línea Germinal , Humanos , Proteínas Inhibidoras de la Apoptosis , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Ratones Mutantes , Ratones Desnudos , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Interferencia de ARN , Proteínas Represoras , Resveratrol , Sirtuina 1 , Estilbenos/farmacología , Survivin
3.
Proc Natl Acad Sci U S A ; 110(33): 13546-51, 2013 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-23898190

RESUMEN

Sirtuin 2 (SIRT2) is a sirtuin family deacetylase that directs acetylome signaling, protects genome integrity, and is a murine tumor suppressor. We show that SIRT2 directs replication stress responses by regulating the activity of cyclin-dependent kinase 9 (CDK9), a protein required for recovery from replication arrest. SIRT2 deficiency results in replication stress sensitivity, impairment in recovery from replication arrest, spontaneous accumulation of replication protein A to foci and chromatin, and a G2/M checkpoint deficit. SIRT2 interacts with and deacetylates CDK9 at lysine 48 in response to replication stress in a manner that is partially dependent on ataxia telangiectasia and Rad3 related (ATR) but not cyclin T or K, thereby stimulating CDK9 kinase activity and promoting recovery from replication arrest. Moreover, wild-type, but not acetylated CDK9, alleviates the replication stress response impairment of SIRT2 deficiency. Collectively, our results define a function for SIRT2 in regulating checkpoint pathways that respond to replication stress through deacetylation of CDK9, providing insight into how SIRT2 maintains genome integrity and a unique mechanism by which SIRT2 may function, at least in part, as a tumor suppressor protein.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Quinasa 9 Dependiente de la Ciclina/metabolismo , Replicación del ADN/fisiología , Sirtuina 2/metabolismo , Acetilación , Animales , Western Blotting , Línea Celular , Cromatografía Liquida , Ensayo de Unidades Formadoras de Colonias , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Proteína de Replicación A/metabolismo , Espectrometría de Masas en Tándem
4.
J Biol Chem ; 289(35): 24202-14, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25006250

RESUMEN

Drug resistance and cancer metastasis are two major problems in cancer research. During a course of therapeutic treatment in Brca1-associated tumors, we found that breast cancer stem cells (CSCs) exhibit an intrinsic ability to metastasize and acquire drug resistance through distinct signaling pathways. Microarray analysis indicated that the cytoskeletal remodeling pathway was differentially regulated in CSCs, and this was further evidenced by the inhibitory role of reagents that impair this pathway in the motility of cancer cells. We showed that cisplatin treatment, although initially inhibiting cancer growth, preventing metastasis through blocking cytoskeletal remodeling, and retarding CSC motility, eventually led to drug resistance associated with a marked increase in the number of CSCs. This event was at least partially attributed to the activation of PI3K signaling, and it could be significantly inhibited by co-treatment with rapamycin. These results provide strong evidence that cytoskeletal rearrangement and PI3K/AKT signaling play distinct roles in mediating CSC mobility and viability, respectively, and blocking both pathways synergistically may inhibit primary and metastatic cancer growth.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Cisplatino/farmacología , Inhibidores Enzimáticos/farmacología , Genes BRCA1 , Metástasis de la Neoplasia/prevención & control , Inhibidores de las Quinasa Fosfoinosítidos-3 , Animales , Resistencia a Antineoplásicos , Citometría de Flujo , Humanos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Análisis de Secuencia por Matrices de Oligonucleótidos
5.
Biochem J ; 454(2): 249-57, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23767918

RESUMEN

Sirt3 (sirtuin 3) is an NAD-dependent deacetylase localized to mitochondria. Sirt3 expression is increased in mouse muscle and liver by starvation, which could protect against the starvation-dependent increase in oxidative stress and protein damage. Damaged proteins and organelles depend on autophagy for removal and this is critical for cell survival, but the role of Sirt3 is unclear. To examine this, we used Sirt3-KO (knockout) mouse embryonic fibroblast cells, and found that, under basal conditions, Sirt3-KO cells exhibited increased autophagy flux compared with WT (wild-type) cells. In response to nutrient deprivation, both WT and KO cells exhibited increased basal and ATP-linked mitochondrial respiration, indicating an increased energy demand. Both cells exhibited lower levels of phosphorylated mTOR (mammalian target of rapamycin) and higher autophagy flux, with KO cells exhibiting lower maximal mitochondrial respiration and reserve capacity, and higher levels of autophagy than WT cells. KO cells exhibit higher phospho-JNK (c-Jun N-terminal kinase) and phospho-c-Jun than WT cells under starvation conditions. However, inhibition of JNK activity in Sirt3-KO cells did not affect LC3-I (light chain 3-I) and LC3-II levels, indicating that Sirt3-regulated autophagy is independent of the JNK pathway. Caspase 3 activation and cell death are significantly higher in Sirt3-KO cells compared with WT cells in response to nutrient deprivation. Inhibition of autophagy by chloroquine exacerbated cell death in both WT and Sirt3-KO cells, and by 3-methyadenine exacerbated cell death in Sirt3-KO cells. These data suggest that nutrient deprivation-induced autophagy plays a protective role in cell survival, and Sirt3 decreases the requirement for enhanced autophagy and improves cellular bioenergetics.


Asunto(s)
Autofagia , Metabolismo Energético , Fibroblastos/metabolismo , Sistema de Señalización de MAP Quinasas , Fosforilación Oxidativa , Sirtuina 3/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Caspasa 3/metabolismo , Células Cultivadas , Medios de Cultivo/metabolismo , Embrión de Mamíferos/citología , Metabolismo Energético/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Sirtuina 3/genética , Serina-Treonina Quinasas TOR/metabolismo , Moduladores de Tubulina/farmacología
6.
Commun Med (Lond) ; 4(1): 22, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378783

RESUMEN

BACKGROUND: Understanding why some triple-negative breast cancer (TNBC) patients respond poorly to existing therapies while others respond well remains a challenge. This study aims to understand the potential underlying mechanisms distinguishing early-stage TNBC tumors that respond to clinical intervention from non-responders, as well as to identify clinically viable therapeutic strategies, specifically for TNBC patients who may not benefit from existing therapies. METHODS: We conducted retrospective bioinformatics analysis of historical gene expression datasets to identify a group of genes whose expression levels in early-stage tumors predict poor clinical outcomes in TNBC. In vitro small-molecule screening, genetic manipulation, and drug treatment in syngeneic mouse models of TNBC were utilized to investigate potential therapeutic strategies and elucidate mechanisms of drug action. RESULTS: Our bioinformatics analysis reveals a robust association between increased expression of immunosuppressive cytokine S100A8/A9 in early-stage tumors and subsequent disease progression in TNBC. A targeted small-molecule screen identifies PIM kinase inhibitors as capable of decreasing S100A8/A9 expression in multiple cell types, including TNBC and immunosuppressive myeloid cells. Combining PIM inhibition and immune checkpoint blockade induces significant antitumor responses, especially in otherwise resistant S100A8/A9-high PD-1/PD-L1-positive tumors. Notably, serum S100A8/A9 levels mirror those of tumor S100A8/A9 in a syngeneic mouse model of TNBC. CONCLUSIONS: Our data propose S100A8/A9 as a potential predictive and pharmacodynamic biomarker in clinical trials evaluating combination therapy targeting PIM and immune checkpoints in TNBC. This work encourages the development of S100A8/A9-based liquid biopsy tests for treatment guidance.


Breast cancer is a complex disease, and not all patients respond well to existing treatments. In this study, we sought to understand why some patients with a specific type of breast cancer called triple-negative breast cancer respond poorly to current therapies. We also aimed to identify new treatments for these patients. We analyzed genetic data from breast cancer patients and identified a factor called S100A8/A9, which is linked to poor outcomes in early-stage cancer. We tested drugs that can reduce the levels of this factor in tumors and found promising results, especially when combined with another treatment called immunotherapy. Our findings suggest that S100A8/A9 could help predict how patients will respond to treatments, potentially leading to better therapies in the future.

7.
Curr Opin Oncol ; 25(6): 630-6, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24048020

RESUMEN

PURPOSE OF REVIEW: The purpose of this review is to highlight recent studies on mammalian sirtuins that coordinately regulate cellular metabolic homeostasis upon fasting and to summarize the beneficial effects of fasting on carcinogenesis and cancer therapy. RECENT FINDINGS: Recent studies have demonstrated that fasting may protect normal cells and mice from the metabolic conditions that are harmful as well as decrease the incidence of carcinogenesis. Fasting could also slow the tumor growth and augment the efficacy of certain systemic agents/chemotherapy drugs in various cancers. The mechanism behind this proposed idea may be due to, at least in some part, the metabolic regulation by Sirtuin family proteins whose functions are involved in specific aspects of longevity, stress response and metabolism. Sirtuins, particularly SIRT1 and SIRT3, can be activated by fasting and further exhibit their effects in insulin response, antioxidant defense, and glycolysis. Therefore, sirtuins may have anticancer effects by shifting metabolism to a less proliferative cell phenotype as well as less prone to oxidative stress attack. SUMMARY: The in-depth understanding of the essential role of sirtuins in fasting process may have significant implications in developing a new metabolic diagram of cancer prevention or treatment.


Asunto(s)
Antioxidantes/metabolismo , Metabolismo Energético , Ayuno/metabolismo , Neoplasias/metabolismo , Estrés Oxidativo , Sirtuina 1/metabolismo , Sirtuina 3/metabolismo , Animales , Femenino , Glucosa/metabolismo , Glucólisis , Homeostasis , Humanos , Insulina/metabolismo , Masculino , Neoplasias/prevención & control , Neoplasias/terapia , Receptor Cross-Talk , Sirtuinas/metabolismo
8.
bioRxiv ; 2023 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-37790346

RESUMEN

It remains elusive why some triple-negative breast cancer (TNBC) patients respond poorly to existing therapies while others respond well. Our retrospective analysis of historical gene expression datasets reveals that increased expression of immunosuppressive cytokine S100A8/A9 in early-stage tumors is robustly associated with subsequent disease progression in TNBC. Although it has recently gained recognition as a potential anticancer target, S100A8/A9 has not been integrated into clinical study designs evaluating molecularly targeted therapies. Our small molecule screen has identified PIM kinase inhibitors as capable of decreasing S100A8/A9 expression in multiple cell types, including TNBC and immunosuppressive myeloid cells. Furthermore, combining PIM inhibition and immune checkpoint blockade induces significant antitumor responses, especially in otherwise resistant S100A8/A9-high PD-1/PD-L1-positive tumors. Importantly, serum S100A8/A9 levels mirror those of tumor S100A8/A9 in a syngeneic mouse model of TNBC. Thus, our data suggest that S100A8/A9 could be a predictive and pharmacodynamic biomarker in clinical trials evaluating combination therapy targeting PIM and immune checkpoints in TNBC and encourage the development of S100A8/A9-based liquid biopsy tests.

9.
Hum Genomics ; 5(5): 485-96, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21807603

RESUMEN

The sirtuin family of proteins is categorised as class III histone deacetylases that play complex and important roles in ageing-related pathological conditions such as cancer and the deregulation of metabolism. There are seven members in humans, divided into four classes, and evolutionarily conserved orthologues can be found in most forms of life, including both eukaryotes and prokaryotes. The highly conserved catalytic core domain composed of a large oxidised nicotinamide adenine dinucleotide (NAD+)-binding Rossmann fold subunit suggests that these proteins belong to a family of nutrient-sensing regulators. Along with their function in regulating cellular metabolism in response to stressful conditions, they are implicated in modifying a wide variety of substrates; this increases the complexity of unravelling the interplay of sirtuins and their partners. Over the past few years, all of these new findings have attracted the interest of researchers exploring potential therapeutic implications related to the function of sirtuins. It remains to be elucidated whether, indeed, sirtuins can serve as molecular targets for the treatment of human illnesses.


Asunto(s)
Evolución Molecular , Sirtuinas/fisiología , Dominio Catalítico , Humanos , Longevidad/fisiología , NAD/química , NAD/metabolismo , Neoplasias/metabolismo , Filogenia , Sirtuinas/química , Sirtuinas/genética
10.
Cell Chem Biol ; 29(3): 358-372.e5, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-34525344

RESUMEN

Triple-negative breast cancer (TNBC) is the breast cancer subtype with the poorest clinical outcome. The PIM family of kinases has emerged as a factor that is both overexpressed in TNBC and associated with poor outcomes. Preclinical data suggest that TNBC with an elevated MYC expression is sensitive to PIM inhibition. However, clinical observations indicate that the efficacy of PIM inhibitors as single agents may be limited, suggesting the need for combination therapies. Our screening effort identifies PIM and the 20S proteasome inhibition as the most synergistic combination. PIM inhibitors, when combined with proteasome inhibitors, induce significant antitumor effects, including abnormal accumulation of poly-ubiquitinated proteins, increased proteotoxic stress, and the inability of NRF1 to counter loss in proteasome activity. Thus, the identified combination could represent a rational combination therapy against MYC-overexpressing TNBC that is readily translatable to clinical investigations.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Proliferación Celular , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-pim-1 , Neoplasias de la Mama Triple Negativas/metabolismo
11.
Proc Natl Acad Sci U S A ; 105(38): 14447-52, 2008 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-18794531

RESUMEN

Here, we demonstrate a role for the mitochondrial NAD-dependent deacetylase Sirt3 in the maintenance of basal ATP levels and as a regulator of mitochondrial electron transport. We note that Sirt3(-/-) mouse embryonic fibroblasts have a reduction in basal ATP levels. Reconstitution with wild-type but not a deacetylase-deficient form of Sirt3 restored ATP levels in these cells. Furthermore in wild-type mice, the resting level of ATP correlates with organ-specific Sirt3 protein expression. Remarkably, in mice lacking Sirt3, basal levels of ATP in the heart, kidney, and liver were reduced >50%. We further demonstrate that mitochondrial protein acetylation is markedly elevated in Sirt3(-/-) tissues. In addition, in the absence of Sirt3, multiple components of Complex I of the electron transport chain demonstrate increased acetylation. Sirt3 can also physically interact with at least one of the known subunits of Complex I, the 39-kDa protein NDUFA9. Functional studies demonstrate that mitochondria from Sirt3(-/-) animals display a selective inhibition of Complex I activity. Furthermore, incubation of exogenous Sirt3 with mitochondria can augment Complex I activity. These results implicate protein acetylation as an important regulator of Complex I activity and demonstrate that Sirt3 functions in vivo to regulate and maintain basal ATP levels.


Asunto(s)
Metabolismo Energético , Homeostasis , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Sirtuinas/metabolismo , Acetilación , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Complejo I de Transporte de Electrón/metabolismo , Femenino , Fibroblastos/citología , Células HeLa , Humanos , Masculino , Ratones , Proteínas Mitocondriales/genética , Sirtuina 3 , Sirtuinas/genética
12.
Curr Mol Med ; 8(8): 727-40, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19075671

RESUMEN

Hereditary breast cancers affect women who have an increased risk of developing tumors because of a familial history. In most cases, they can be attributed to mutations in the breast cancer associated gene 1 and 2 (BRCA1 and BRCA2). Recent studies have demonstrated a link between the insulin-like growth factor (IGF) signaling pathway and familial breast cancer incidence. IGF and IGF receptors represent a family of biological growth factors and transducers, which have been involved in both physiological and pathological processes. It has been shown that BRCA1 regulates expression of several members of the IGF family. Here, we will examine our understanding of the functions of IGF/IGF-receptor signaling, the development of new inhibitors of this pathway and the related mechanisms of familial breast cancer formation.


Asunto(s)
Neoplasias de la Mama/terapia , Somatomedinas/fisiología , Animales , Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Femenino , Genes BRCA1 , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/fisiopatología , Neoplasias Mamarias Experimentales/terapia , Ratones , Modelos Biológicos , Inhibidores de Proteínas Quinasas/uso terapéutico , ARN Interferente Pequeño/genética , Receptores de Somatomedina/antagonistas & inhibidores , Receptores de Somatomedina/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Somatomedinas/antagonistas & inhibidores
13.
Front Oncol ; 9: 1462, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31970087

RESUMEN

Calorie restriction (CR) is considered one of the most robust ways to extend life span and reduce the risk of age-related diseases, including cancer, as shown in many different organisms, whereas opposite effects have been associated with high fat diets (HFDs). Despite the proven contribution of sirtuins in mediating the effects of CR in longevity, the involvement of these nutrient sensors, specifically, in the diet-induced effects on tumorigenesis has yet to be elucidated. Previous studies focusing on SIRT1, do not support a critical role for this sirtuin family member in CR-mediated cancer prevention. However, the contribution of other family members which exhibit strong deacetylase activity is unexplored. To fill this gap, we aimed at investigating the role of SIRT2 and SIRT3 in mediating the anti and pro-tumorigenic effect of CR and HFD, respectively. Our results provide strong evidence supporting distinct, context-dependent roles played by these two family members. SIRT2 is indispensable for the protective effect of CR against tumorigenesis. On the contrary, SIRT3 exhibited oncogenic properties in the context of HFD-induced tumorigenesis, suggesting that SIRT3 inhibition may mitigate the cancer-promoting effects of HFD. Given the different functions regulated by SIRT2 and SIRT3, unraveling downstream targets/pathways involved may provide opportunities to develop new strategies for cancer prevention.

14.
Free Radic Biol Med ; 126: 358-371, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30114477

RESUMEN

Previous studies have shown that SIRT2 plays a role in mitosis through deacetylating specific downstream targets. However, the upstream regulation of SIRT2 activity has been relatively unexplored. In this study, we provide evidence that NAD(P)H:quinone oxidoreductase 1 (NQO1) interacts with and activates SIRT2 in an NAD-dependent manner. Strong protein-protein interaction and co-localization of the two proteins during mitosis is required to maintain an active NQO1-SIRT2 axis which is critical for successful completion of mitosis. This is evident by the observed delay in mitotic exit in cells upon NQO1 inhibition. Mechanistically, this phenotype can be explained by the decrease in APC/C complex activity resulting from decreased SIRT2 deacetylation activity. Furthermore, we show that this newly established role of NQO1 has an impact on how cancer cells may respond to mitotic stress. In this regard, both pharmacologic and genetic NQO1 inhibition increases sensitivity to anti-mitotic drugs functioning as microtubule poisons by inducing mitotic arrest and allowing cells to accumulate cell death signals. Therefore, the significant prognostic value of NQO1 in predicting outcome of cancer patients might be explained in part due to the functional contribution of NQO1-SIRT2 axis to mitotic stress. Altogether, this novel mechanism of action further supports the pleiotropic biological effects exerted by NQO1 in addition to its antioxidant function and it might provide the basis for expanding the therapeutic potential of NQO1 inhibition towards increasing sensitivity to standard treatments.


Asunto(s)
Antioxidantes/metabolismo , Mitosis/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , Neoplasias/genética , Sirtuina 2/genética , Muerte Celular/genética , Proliferación Celular/genética , Humanos , Células MCF-7 , Microtúbulos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Mapas de Interacción de Proteínas/genética , Transducción de Señal/genética
15.
Free Radic Biol Med ; 123: 116-124, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29778462

RESUMEN

Impaired bioenergetics and oxidative damage in the mitochondria are implicated in the etiology of temporal lobe epilepsy, and hyperacetylation of mitochondrial proteins has recently emerged as a critical negative regulator of mitochondrial functions. However, the roles of mitochondrial acetylation and activity of the primary mitochondrial deacetylase, SIRT3, have not been explored in acquired epilepsy. We investigated changes in mitochondrial acetylation and SIRT3 activity in the development of chronic epilepsy in the kainic acid rat model of TLE. Hippocampal measurements were made at 48 h, 1 week and 12 weeks corresponding to the acute, latent and chronic stages of epileptogenesis. Assessment of hippocampal bioenergetics demonstrated a ≥ 27% decrease in the ATP/ADP ratio at all phases of epileptogenesis (p < 0.05), whereas cellular NAD+ levels were decreased by ≥ 41% in the acute and latent time points (p < 0.05), but not in chronically epileptic rats. In spontaneously epileptic rats, we found decreased protein expression of SIRT3 and a 60% increase in global mitochondrial acetylation, as well as enhanced acetylation of the known SIRT3 substrates MnSOD, Ndufa9 of Complex I and IDH2 (all p < 0.05), suggesting SIRT3 dysfunction in chronic epilepsy. Mass spectrometry-based acetylomics investigation of hippocampal mitochondria demonstrated a 79% increase in unique acetylated proteins from rats in the chronic phase vs. controls. Pathway analysis identified numerous mitochondrial bioenergetic pathways affected by mitochondrial acetylation. These results suggest SIRT3 dysfunction and aberrant protein acetylation may contribute to mitochondrial dysfunction in chronic epilepsy.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/metabolismo , Agonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/metabolismo , Ácido Kaínico/toxicidad , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Acetilación , Animales , Metabolismo Energético , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/patología , Hipocampo/patología , Masculino , Mitocondrias/patología , Ratas , Ratas Sprague-Dawley , Sirtuinas/metabolismo
16.
Aging Cell ; 16(6): 1208-1218, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28994177

RESUMEN

Sirtuins are stress-responsive proteins that direct various post-translational modifications (PTMs) and as a result, are considered to be master regulators of several cellular processes. They are known to both extend lifespan and regulate spontaneous tumor development. As both aging and cancer are associated with altered stem cell function, the possibility that the involvement of sirtuins in these events is mediated by their roles in stem cells is worthy of investigation. Research to date suggests that the individual sirtuin family members can differentially regulate embryonic, hematopoietic as well as other adult stem cells in a tissue- and cell type-specific context. Sirtuin-driven regulation of both cell differentiation and signaling pathways previously involved in stem cell maintenance has been described where downstream effectors involved determine the biological outcome. Similarly, diverse roles have been reported in cancer stem cells (CSCs), depending on the tissue of origin. This review highlights the current knowledge which places sirtuins at the intersection of stem cells, aging, and cancer. By outlining the plethora of stem cell-related roles for individual sirtuins in various contexts, our purpose was to provide an indication of their significance in relation to cancer and aging, as well as to generate a clearer picture of their therapeutic potential. Finally, we propose future directions which will contribute to the better understanding of sirtuins, thereby further unraveling the full repertoire of sirtuin functions in both normal stem cells and CSCs.


Asunto(s)
Neoplasias/genética , Sirtuinas/genética , Células Madre/metabolismo , Envejecimiento , Diferenciación Celular , Humanos , Sirtuinas/metabolismo
17.
J Clin Invest ; 127(4): 1505-1516, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28287409

RESUMEN

SIRT2 is a cytoplasmic sirtuin that plays a role in various cellular processes, including tumorigenesis, metabolism, and inflammation. Since these processes require iron, we hypothesized that SIRT2 directly regulates cellular iron homeostasis. Here, we have demonstrated that SIRT2 depletion results in a decrease in cellular iron levels both in vitro and in vivo. Mechanistically, we determined that SIRT2 maintains cellular iron levels by binding to and deacetylating nuclear factor erythroid-derived 2-related factor 2 (NRF2) on lysines 506 and 508, leading to a reduction in total and nuclear NRF2 levels. The reduction in nuclear NRF2 leads to reduced ferroportin 1 (FPN1) expression, which in turn results in decreased cellular iron export. Finally, we observed that Sirt2 deletion reduced cell viability in response to iron deficiency. Moreover, livers from Sirt2-/- mice had decreased iron levels, while this effect was reversed in Sirt2-/- Nrf2-/- double-KO mice. Taken together, our results uncover a link between sirtuin proteins and direct control over cellular iron homeostasis via regulation of NRF2 deacetylation and stability.


Asunto(s)
Hierro/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Procesamiento Proteico-Postraduccional , Sirtuina 2/fisiología , Acetilación , Animales , Proteínas de Transporte de Catión/metabolismo , Epigénesis Genética , Expresión Génica , Células HEK293 , Células Hep G2 , Homeostasis , Humanos , Hígado/metabolismo , Ratones Noqueados , Estabilidad Proteica , Activación Transcripcional
18.
J Vis Exp ; (108): 53563, 2016 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-26966987

RESUMEN

Acetylation has emerged as an important post-translational modification (PTM) regulating a plethora of cellular processes and functions. This is further supported by recent findings in high-resolution mass spectrometry based proteomics showing that many new proteins and sites within these proteins can be acetylated. However the identity of the enzymes regulating these proteins and sites is often unknown. Among these enzymes, sirtuins, which belong to the class III histone lysine deacetylases, have attracted great interest as enzymes regulating the acetylome under different physiological or pathophysiological conditions. Here we describe methods to link SIRT2, the cytoplasmic sirtuin, with its substrates including both in vitro and in vivo deacetylation assays. These assays can be applied in studies focused on other members of the sirtuin family to unravel the specific role of sirtuins and are necessary in order to establish the regulatory interplay of specific deacetylases with their substrates as a first step to better understand the role of protein acetylation. Furthermore, such assays can be used to distinguish functional acetylation sites on a protein from what may be non-regulatory acetylated lysines, as well as to examine the interplay between a deacetylase and its substrate in a physiological context.


Asunto(s)
Acetilación , Sirtuinas/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Lisina/química , Espectrometría de Masas , Procesamiento Proteico-Postraduccional/fisiología , Proteínas/metabolismo , Proteómica
19.
Cancer Res ; 76(13): 3802-12, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27197174

RESUMEN

Sirtuins participate in sensing nutrient availability and directing metabolic activity to match energy needs with energy production and consumption. However, the pivotal targets for sirtuins in cancer are mainly unknown. In this study, we identify the M2 isoform of pyruvate kinase (PKM2) as a critical target of the sirtuin SIRT2 implicated in cancer. PKM2 directs the synthesis of pyruvate and acetyl-CoA, the latter of which is transported to mitochondria for use in the Krebs cycle to generate ATP. Enabled by a shotgun mass spectrometry analysis founded on tissue culture models, we identified a candidate SIRT2 deacetylation target at PKM2 lysine 305 (K305). Biochemical experiments including site-directed mutants that mimicked constitutive acetylation suggested that acetylation reduced PKM2 activity by preventing tetramerization to the active enzymatic form. Notably, ectopic overexpression of a deacetylated PKM2 mutant in Sirt2-deficient mammary tumor cells altered glucose metabolism and inhibited malignant growth. Taken together, our results argued that loss of SIRT2 function in cancer cells reprograms their glycolytic metabolism via PKM2 regulation, partially explaining the tumor-permissive phenotype of mice lacking Sirt2 Cancer Res; 76(13); 3802-12. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Glucosa/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Recurrencia Local de Neoplasia/patología , Sirtuina 2/fisiología , Hormonas Tiroideas/química , Hormonas Tiroideas/metabolismo , Acetilación , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Glucólisis , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Noqueados , Recurrencia Local de Neoplasia/metabolismo , Estadificación de Neoplasias , Pronóstico , Análisis de Matrices Tisulares , Células Tumorales Cultivadas , Proteínas de Unión a Hormona Tiroide
20.
Oncotarget ; 7(49): 80336-80349, 2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27637077

RESUMEN

The observation that cellular transformation depends on breaching a crucial KRAS activity threshold, along with the finding that only a small percentage of cellsharboring KRAS mutations are transformed, support the idea that additional, not fully uncovered, regulatory mechanisms may contribute to KRAS activation. Here we report that KrasG12D mice lacking Sirt2 show an aggressive tumorigenic phenotype as compared to KrasG12D mice. This phenotype includes increased proliferation, KRAS acetylation, and activation of RAS downstream signaling markers. Mechanistically, KRAS K147 is identified as a novel SIRT2-specific deacetylation target by mass spectrometry, whereas its acetylation status directly regulates KRAS activity, ultimately exerting an impact on cellular behavior as revealed by cell proliferation, colony formation, and tumor growth. Given the significance of KRAS activity as a driver in tumorigenesis, identification of K147 acetylation as a novel post-translational modification directed by SIRT2 in vivo may provide a better understanding of the mechanistic link regarding the crosstalk between non-genetic and genetic factors in KRAS driven tumors.


Asunto(s)
Adenocarcinoma/enzimología , Transformación Celular Neoplásica/metabolismo , Eliminación de Gen , Neoplasias Pulmonares/enzimología , Neoplasias Pancreáticas/enzimología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sirtuina 2/deficiencia , Acetilación , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HCT116 , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Lisina , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Mutación , Células 3T3 NIH , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal , Sirtuina 2/genética , Factores de Tiempo , Carga Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA