Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Sci Adv ; 10(20): eadj5942, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758779

RESUMEN

Acetyl-CoA synthetase short-chain family member 1 (ACSS1) uses acetate to generate mitochondrial acetyl-CoA and is regulated by deacetylation by sirtuin 3. We generated an ACSS1-acetylation (Ac) mimic mouse, where lysine-635 was mutated to glutamine (K635Q). Male Acss1K635Q/K635Q mice were smaller with higher metabolic rate and blood acetate and decreased liver/serum ATP and lactate levels. After a 48-hour fast, Acss1K635Q/K635Q mice presented hypothermia and liver aberrations, including enlargement, discoloration, lipid droplet accumulation, and microsteatosis, consistent with nonalcoholic fatty liver disease (NAFLD). RNA sequencing analysis suggested dysregulation of fatty acid metabolism, cellular senescence, and hepatic steatosis networks, consistent with NAFLD. Fasted Acss1K635Q/K635Q mouse livers showed increased fatty acid synthase (FASN) and stearoyl-CoA desaturase 1 (SCD1), both associated with NAFLD, and increased carbohydrate response element-binding protein binding to Fasn and Scd1 enhancer regions. Last, liver lipidomics showed elevated ceramide, lysophosphatidylethanolamine, and lysophosphatidylcholine, all associated with NAFLD. Thus, we propose that ACSS1-K635-Ac dysregulation leads to aberrant lipid metabolism, cellular senescence, and NAFLD.


Asunto(s)
Senescencia Celular , Mitocondrias , Enfermedad del Hígado Graso no Alcohólico , Estearoil-CoA Desaturasa , Animales , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Ratones , Senescencia Celular/genética , Acetilación , Mitocondrias/metabolismo , Estearoil-CoA Desaturasa/metabolismo , Estearoil-CoA Desaturasa/genética , Masculino , Acetato CoA Ligasa/metabolismo , Acetato CoA Ligasa/genética , Técnicas de Sustitución del Gen , Hígado/metabolismo , Hígado/patología , Metabolismo de los Lípidos , Sirtuina 3/metabolismo , Sirtuina 3/genética , Modelos Animales de Enfermedad , Coenzima A Ligasas , Acido Graso Sintasa Tipo I
2.
Sci Adv ; 10(20): eado1463, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758782

RESUMEN

A ketogenic diet (KD) is a high-fat, low-carbohydrate diet that leads to the generation of ketones. While KDs improve certain health conditions and are popular for weight loss, detrimental effects have also been reported. Here, we show mice on two different KDs and, at different ages, induce cellular senescence in multiple organs, including the heart and kidney. This effect is mediated through adenosine monophosphate-activated protein kinase (AMPK) and inactivation of mouse double minute 2 (MDM2) by caspase-2, leading to p53 accumulation and p21 induction. This was established using p53 and caspase-2 knockout mice and inhibitors to AMPK, p21, and caspase-2. In addition, senescence-associated secretory phenotype biomarkers were elevated in serum from mice on a KD and in plasma samples from patients on a KD clinical trial. Cellular senescence was eliminated by a senolytic and prevented by an intermittent KD. These results have important clinical implications, suggesting that the effects of a KD are contextual and likely require individual optimization.


Asunto(s)
Senescencia Celular , Dieta Cetogénica , Ratones Noqueados , Proteína p53 Supresora de Tumor , Animales , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Ratones , Humanos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Masculino , Especificidad de Órganos
3.
Antioxidants (Basel) ; 11(4)2022 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-35453320

RESUMEN

The loss and/or dysregulation of several cellular and mitochondrial antioxidants' expression or enzymatic activity, which leads to the aberrant physiological function of these proteins, has been shown to result in oxidative damage to cellular macromolecules. In this regard, it has been surmised that the disruption of mitochondrial networks responsible for maintaining normal metabolism is an established hallmark of cancer and a novel mechanism of therapy resistance. This altered metabolism leads to aberrant accumulation of reactive oxygen species (ROS), which, under specific physiological conditions, leads to a potential tumor-permissive cellular environment. In this regard, it is becoming increasingly clear that the loss or disruption of mitochondrial oxidant scavenging enzymes may be, in specific tumors, either an early event in transformation or exhibit tumor-promoting properties. One example of such an antioxidant enzyme is manganese superoxide dismutase (MnSOD, also referred to as SOD2), which detoxifies superoxide, a ROS that has been shown, when its normal physiological levels are disrupted, to lead to oncogenicity and therapy resistance. Here, we will also discuss how the acetylation of MnSOD leads to a change in detoxification function that leads to a cellular environment permissive for the development of lineage plasticity-like properties that may be one mechanism leading to tumorigenic and therapy-resistant phenotypes.

4.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202229

RESUMEN

Alveolar epithelial cell (AEC) mitochondrial (mt) DNA damage and fibrotic monocyte-derived alveolar macrophages (Mo-AMs) are implicated in the pathobiology of pulmonary fibrosis. We showed that sirtuin 3 (SIRT3), a mitochondrial protein regulating cell fate and aging, is deficient in the AECs of idiopathic pulmonary fibrosis (IPF) patients and that asbestos- and bleomycin-induced lung fibrosis is augmented in Sirt3 knockout (Sirt3-/-) mice associated with AEC mtDNA damage and intrinsic apoptosis. We determined whether whole body transgenic SIRT3 overexpression (Sirt3Tg) protects mice from asbestos-induced pulmonary fibrosis by mitigating lung mtDNA damage and Mo-AM recruitment. Crocidolite asbestos (100 µg/50 µL) or control was instilled intratracheally in C57Bl6 (Wild-Type) mice or Sirt3Tg mice, and at 21 d lung fibrosis (histology, fibrosis score, Sircol assay) and lung Mo-AMs (flow cytometry) were assessed. Compared to controls, Sirt3Tg mice were protected from asbestos-induced pulmonary fibrosis and had diminished lung mtDNA damage and Mo-AM recruitment. Further, pharmacologic SIRT3 inducers (i.e., resveratrol, viniferin, and honokiol) each diminish oxidant-induced AEC mtDNA damage in vitro and, in the case of honokiol, protection occurs in a SIRT3-dependent manner. We reason that SIRT3 preservation of AEC mtDNA is a novel therapeutic focus for managing patients with IPF and other types of pulmonary fibrosis.


Asunto(s)
Amianto/efectos adversos , Daño del ADN , Expresión Génica , Fibrosis Pulmonar Idiopática/etiología , Mitocondrias/genética , Monocitos/metabolismo , Sirtuina 3/genética , Animales , Biomarcadores , ADN Mitocondrial , Modelos Animales de Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Inmunohistoquímica , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Monocitos/inmunología , Monocitos/patología , Estrés Oxidativo , Sirtuina 3/metabolismo
5.
J Natl Cancer Inst ; 113(10): 1285-1298, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33792717

RESUMEN

Cellular senescence is an essential tumor suppressive mechanism that prevents the propagation of oncogenically activated, genetically unstable, and/or damaged cells. Induction of tumor cell senescence is also one of the underlying mechanisms by which cancer therapies exert antitumor activity. However, an increasing body of evidence from preclinical studies demonstrates that radiation and chemotherapy cause accumulation of senescent cells (SnCs) both in tumor and normal tissue. SnCs in tumors can, paradoxically, promote tumor relapse, metastasis, and resistance to therapy, in part, through expression of the senescence-associated secretory phenotype. In addition, SnCs in normal tissue can contribute to certain radiation- and chemotherapy-induced side effects. Because of its multiple roles, cellular senescence could serve as an important target in the fight against cancer. This commentary provides a summary of the discussion at the National Cancer Institute Workshop on Radiation, Senescence, and Cancer (August 10-11, 2020, National Cancer Institute, Bethesda, MD) regarding the current status of senescence research, heterogeneity of therapy-induced senescence, current status of senotherapeutics and molecular biomarkers, a concept of "one-two punch" cancer therapy (consisting of therapeutics to induce tumor cell senescence followed by selective clearance of SnCs), and its integration with personalized adaptive tumor therapy. It also identifies key knowledge gaps and outlines future directions in this emerging field to improve treatment outcomes for cancer patients.


Asunto(s)
Senescencia Celular , Neoplasias , Biomarcadores , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fenotipo Secretor Asociado a la Senescencia
6.
Int J Biol Sci ; 17(5): 1203-1216, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33867840

RESUMEN

Manganese superoxide dismutase (MnSOD) acetylation (Ac) has been shown to be a key post-translational modification important in the regulation of detoxification activity in various disease models. We have previously demonstrated that MnSOD lysine-68 (K68) acetylation (K68-Ac) leads to a change in function from a superoxide-scavenging homotetramer to a peroxidase-directed monomer. Here, we found that estrogen receptor positive (ER+) breast cancer cell lines (MCF7 and T47D), selected for continuous growth in cisplatin (CDDP) and doxorubicin (DXR), exhibited an increase in MnSOD-K68-Ac. In addition, MnSOD-K68-Ac, as modeled by the expression of a validated acetylation mimic mutant gene (MnSODK68Q ), also led to therapy resistance to CDDP and DXR, altered mitochondrial structure and morphology, and aberrant cellular metabolism. MnSODK68Q expression in mouse embryo fibroblasts (MEFs) induced an in vitro transformation permissive phenotype. Computerized molecular protein dynamics analysis of both MnSOD-K68-Ac and MnSOD-K68Q exhibited a significant change in charge distribution along the α1 and α2 helices, directly adjacent to the Mn2+ binding site, implying that this decrease in surface charge destabilizes tetrameric MnSOD, leading to an enrichment of the monomer. Finally, monomeric MnSOD, as modeled by amber codon substitution to generate MnSOD-K68-Ac or MnSOD-K68Q expression in mammalian cells, appeared to incorporate Fe to maximally induce its peroxidase activity. In summary, these findings may explain the mechanism behind the observed structural and functional change of MnSOD-K68-Ac.


Asunto(s)
Neoplasias de la Mama , Carcinogénesis , Cisplatino/farmacología , Doxorrubicina/farmacología , Mitocondrias , Sirtuinas/metabolismo , Superóxido Dismutasa/metabolismo , Acetilación , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Depuradores de Radicales Libres/metabolismo , Humanos , Inactivación Metabólica , Células MCF-7 , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Procesamiento Proteico-Postraduccional
7.
Proc Natl Acad Sci U S A ; 117(29): 16938-16948, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32616570

RESUMEN

Despite nearly four decades of effort, broad inhibition of oncogenic RAS using small-molecule approaches has proven to be a major challenge. Here we describe the development of a pan-RAS biologic inhibitor composed of the RAS-RAP1-specific endopeptidase fused to the protein delivery machinery of diphtheria toxin. We show that this engineered chimeric toxin irreversibly cleaves and inactivates intracellular RAS at low picomolar concentrations terminating downstream signaling in receptor-bearing cells. Furthermore, we demonstrate in vivo target engagement and reduction of tumor burden in three mouse xenograft models driven by either wild-type or mutant RAS Intracellular delivery of a potent anti-RAS biologic through a receptor-mediated mechanism represents a promising approach to developing RAS therapeutics against a broad array of cancers.


Asunto(s)
Toxina Diftérica/metabolismo , Endopeptidasas/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Proteolisis , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/metabolismo , Animales , Antineoplásicos/uso terapéutico , Células Cultivadas , Toxina Diftérica/química , Toxina Diftérica/genética , Endopeptidasas/química , Endopeptidasas/genética , Femenino , Células HCT116 , Humanos , Masculino , Ratones , Ratones Desnudos , Mutación , Señales de Clasificación de Proteína , Proteínas Recombinantes/uso terapéutico , Proteínas ras/genética
8.
Cell Signal ; 70: 109574, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32084531

RESUMEN

KLF4 plays an important role in orchestrating a variety of cellular events, including cell-fate decision, genome stability and apoptosis. Its deregulation is correlated with human diseases such as breast cancer and gastrointestinal cancer. Results from recent biochemical studies have revealed that KLF4 is tightly regulated by posttranslational modifications. Here we report a new finding that KLF4 orchestrates estrogen receptor signaling and facilitates endocrine resistance. We also uncovered the underlying mechanism that alteration of KLF4 by posttranslational modifications such as phosphorylation and ubiquitylation changes tumor cell response to endocrine therapy drugs. IHC analyses using based on human breast cancer specimens showed the accumulation of KLF4 protein in ER-positive breast cancer tissues. Elevated KLF4 expression significantly correlated with prognosis and endocrine resistance. Our drug screening for suppressing KLF4 protein expression led to identification of Src kinase to be a critical player in modulating KLF4-mediated tamoxifen resistance. Depletion of VHL (von Hippel-Lindau tumor suppressor), a ubiquitin E3 ligase for KLF4, reduces tumor cell sensitivity to tamoxifen. We demonstrated phosphorylation of VHL by Src enhances proteolysis of VHL that in turn leads to upregulation of KLF4 and increases endocrine resistance. Suppression of Src-VHL-KLF4 cascade by Src inhibitor or enhancement of VHL-KLF4 ubiquitination by TAT-KLF4 (371-420AAa) peptides re-sensitizes tamoxifen-resistant breast cancer cells to tamoxifen treatment. Taken together, our findings demonstrate a novel role for KLF4 in modulating endocrine resistance via the Src-VHL-KLF4 axis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Células MCF-7
9.
Am J Cancer Res ; 10(12): 4416-4434, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33415008

RESUMEN

Cisplatin is a potent drug used in about 40% of cancer treatment but also leads to severe deafness in 60-80% of the cases. Although the mechanism is known to be related to the accumulation of reactive oxygen species (ROS), no drug or FDA approved treatment is currently available to prevent cisplatin ototoxicity. With this study, we show for the first time that honokiol (HNK), a pleiotropic poly-phenol prevents cisplatin-induced hearing loss. HNK also improves the wellbeing of the mice during the treatment, determined by the increase in the number of surviving animals. In a transgenic tumor mouse model, HNK does not hinder cisplatin's antitumor effect. The mechanism is related to the activation of sirtuin 3, a deacetylase in mitochondria essential for ROS detoxification. We expect a paradigm shift in cisplatin chemotherapy based on the current study and future clinical trials, where honokiol is applied to reduce side effects including hearing loss.

10.
Proc Natl Acad Sci U S A ; 116(47): 23534-23541, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31591207

RESUMEN

Mitochondrial superoxide dismutase (SOD2) suppresses tumor initiation but promotes invasion and dissemination of tumor cells at later stages of the disease. The mechanism of this functional switch remains poorly defined. Our results indicate that as SOD2 expression increases acetylation of lysine 68 ensues. Acetylated SOD2 promotes hypoxic signaling via increased mitochondrial reactive oxygen species (mtROS). mtROS, in turn, stabilize hypoxia-induced factor 2α (HIF2α), a transcription factor upstream of "stemness" genes such as Oct4, Sox2, and Nanog. In this sense, our findings indicate that SOD2K68Ac and mtROS are linked to stemness reprogramming in breast cancer cells via HIF2α signaling. Based on these findings we propose that, as tumors evolve, the accumulation of SOD2K68Ac turns on a mitochondrial pathway to stemness that depends on HIF2α and may be relevant for the progression of breast cancer toward poor outcomes.


Asunto(s)
Neoplasias de la Mama/patología , Autorrenovación de las Células/fisiología , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/fisiología , Superóxido Dismutasa/fisiología , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Neoplasias de la Mama/metabolismo , Reprogramación Celular , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Peróxido de Hidrógeno/metabolismo , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/enzimología , Invasividad Neoplásica , Proteínas de Neoplasias/química , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Superóxido Dismutasa/química
11.
Nat Commun ; 10(1): 2399, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31160585

RESUMEN

Manganese superoxide dismutase (MnSOD) functions as a tumor suppressor; however, once tumorigenesis occurs, clinical data suggest MnSOD levels correlate with more aggressive human tumors, implying a potential dual function of MnSOD in the regulation of metabolism. Here we show, using in vitro transformation and xenograft growth assays that the MnSOD-K68 acetylation (Ac) mimic mutant (MnSODK68Q) functions as a tumor promoter. Interestingly, in various breast cancer and primary cell types the expression of MnSODK68Q is accompanied with a change of MnSOD's stoichiometry from a known homotetramer complex to a monomeric form. Biochemical experiments using the MnSOD-K68Q Ac-mimic, or physically K68-Ac (MnSOD-K68-Ac), suggest that these monomers function as a peroxidase, distinct from the established MnSOD superoxide dismutase activity. MnSODK68Q expressing cells exhibit resistance to tamoxifen (Tam) and cells selected for Tam resistance exhibited increased K68-Ac and monomeric MnSOD. These results suggest a MnSOD-K68-Ac metabolic pathway for Tam resistance, carcinogenesis and tumor progression.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Resistencia a Antineoplásicos/genética , Superóxido Dismutasa/genética , Acetilación , Animales , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Técnicas In Vitro , Lisina/metabolismo , Células MCF-7 , Ratones , Mutación , Trasplante de Neoplasias , Peroxidasa/metabolismo , Estructura Cuaternaria de Proteína/genética , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Tamoxifeno/uso terapéutico , Proteínas Supresoras de Tumor
12.
Cancer Res ; 79(7): 1295-1296, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30936074

RESUMEN

In this issue of Cancer Research, Ranoa and colleagues report on the role of STING (stimulator of IFN genes, TMEM173) in regulating critical tumor cell-intrinsic functions including cell-cycle progression, chromosomal stability, and cellular response to therapeutic ionizing radiation. The authors used multiple methods including RNA expression profiling, molecular and biochemical techniques, cell biology, and reagents from genetically modified murine models to test their hypothesis that downregulating the STING pathway in cancer cells promotes cellular transformation through accumulation of chromosomal instability and premature progression of the cell cycle. Their findings demonstrate that STING is a tumor suppressor that inhibits cell proliferation by restricting entry to mitosis as well as protecting cells against aneuploidy. These findings significantly advance our understanding of the role of STING as a tumor gate keeper.See related article by Ranoa et al., p. 1465.


Asunto(s)
Proteínas de la Membrana/genética , Neoplasias/genética , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Inestabilidad Cromosómica , Ratones
13.
Cell Metab ; 29(2): 443-456.e5, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30595481

RESUMEN

During wound injury, efferocytosis fills the macrophage with a metabolite load nearly equal to the phagocyte itself. A timely question pertains to how metabolic phagocytic signaling regulates the signature anti-inflammatory macrophage response. Here we report the metabolome of activated macrophages during efferocytosis to reveal an interleukin-10 (IL-10) cytokine escalation that was independent of glycolysis yet bolstered by apoptotic cell fatty acids and mitochondrial ß-oxidation, the electron transport chain, and heightened coenzyme NAD+. Loss of IL-10 due to mitochondrial complex III defects was remarkably rescued by adding NAD+ precursors. This activated a SIRTUIN1 signaling cascade, largely independent of ATP, that culminated in activation of IL-10 transcription factor PBX1. Il-10 activation by the respiratory chain was also important in vivo, as efferocyte mitochondrial dysfunction led to cardiac rupture after myocardial injury. These findings highlight a new paradigm whereby macrophages leverage efferocytic metabolites and electron transport for anti-inflammatory reprogramming that culminates in organ repair.


Asunto(s)
Ácidos Grasos/metabolismo , Interleucina-10/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo , NAD/metabolismo , Animales , Citofagocitosis , Transporte de Electrón , Humanos , Inflamación/metabolismo , Células Jurkat , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Cicatrización de Heridas
14.
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
15.
PLoS One ; 13(12): e0208634, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30533032

RESUMEN

The NAD+-dependent deacetylase SIRT2 is unique amongst sirtuins as it is effective in the cytosol, as well as the mitochondria. Defining the role of cytosolic acetylation state in specific tissues is difficult since even physiological effects at the whole body level are unknown. We hypothesized that genetic SIRT2 knockout (KO) would lead to impaired insulin action, and that this impairment would be worsened in HF fed mice. Insulin sensitivity was tested using the hyperinsulinemic-euglycemic clamp in SIRT2 KO mice and WT littermates. SIRT2 KO mice exhibited reduced skeletal muscle insulin-induced glucose uptake compared to lean WT mice, and this impairment was exacerbated in HF SIRT2 KO mice. Liver insulin sensitivity was unaffected in lean SIRT2 KO mice. However, the insulin resistance that accompanies HF-feeding was worsened in SIRT2 KO mice. It was notable that the effects of SIRT2 KO were largely disassociated from cytosolic acetylation state, but were closely linked to acetylation state in the mitochondria. SIRT2 KO led to an increase in body weight that was due to increased food intake in HF fed mice. In summary, SIRT2 deletion in vivo reduces muscle insulin sensitivity and contributes to liver insulin resistance by a mechanism that is unrelated to cytosolic acetylation state. Mitochondrial acetylation state and changes in feeding behavior that result in increased body weight correspond to the deleterious effects of SIRT2 KO on insulin action.


Asunto(s)
Dieta Alta en Grasa , Resistencia a la Insulina , Sirtuina 2/genética , Acetilación/efectos de los fármacos , Animales , Metabolismo Energético , Insulina/sangre , Insulina/farmacología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirtuina 2/deficiencia
16.
Sci Rep ; 8(1): 16501, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30405152

RESUMEN

Mice lacking Sirt2 spontaneously develop tumors in multiple organs, as well as when expressed in combination with oncogenic KrasG12D, leading to pancreatic tumors. Here, we report that after caerulein-induced pancreatitis, Sirt2-deficient mice exhibited an increased inflammatory phenotype and delayed pancreatic tissue recovery. Seven days post injury, the pancreas of Sirt2-/- mice display active inflammation, whereas wild-type mice had mostly recovered. In addition, the pancreas from the Sirt2-/- mice exhibited extensive tissue fibrosis, which was still present at six weeks after exposure. The mice lacking Sirt2 also demonstrated an enhanced whole body pro-inflammatory phenotype that was most obvious with increasing age. Importantly, an accumulation of a cell population with spontaneous cancerous KrasG12D mutations was observed in the Sirt2-/- mice that is enhanced in the recovering pancreas after exposure to caerulein. Finally, transcriptome analysis of the pancreas of the Sirt2-/- mice exhibited a pro-inflammatory genomic signature. These results suggest that loss of Sirt2, as well as increased age, enhanced the immune response to pancreatic injury and induced an inflammatory phenotype permissive for the accumulation of cells carrying oncogenic Kras mutations.


Asunto(s)
Ceruletida/efectos adversos , Mutación , Pancreatitis/etiología , Pancreatitis/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Sirtuina 2/genética , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Femenino , Predisposición Genética a la Enfermedad , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Pancreatitis/patología , Regeneración
17.
J Clin Invest ; 128(9): 3682-3691, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30168803

RESUMEN

Mammalian cells use a complex network of redox-dependent processes necessary to maintain cellular integrity during oxidative metabolism, as well as to protect against and/or adapt to stress. The disruption of these redox-dependent processes, including those in the mitochondria, creates a cellular environment permissive for progression to a malignant phenotype and the development of resistance to commonly used anticancer agents. An extension of this paradigm is that when these mitochondrial functions are altered by the events leading to transformation and ensuing downstream metabolic processes, they can be used as molecular biomarkers or targets in the development of new therapeutic interventions to selectively kill and/or sensitize cancer versus normal cells. In this Review we propose that mitochondrial oxidative metabolism is altered in tumor cells, and the central theme of this dysregulation is electron transport chain activity, folate metabolism, NADH/NADPH metabolism, thiol-mediated detoxification pathways, and redox-active metal ion metabolism. It is proposed that specific subgroups of human malignancies display distinct mitochondrial transformative and/or tumor signatures that may benefit from agents that target these pathways.


Asunto(s)
Mitocondrias/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Animales , Femenino , Expresión Génica , Humanos , Masculino , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , NAD/metabolismo , NADP/metabolismo , Neoplasias/genética , Oxidación-Reducción , Estrés Oxidativo , Transporte de Proteínas , Transducción de Señal , Sirtuinas/metabolismo
18.
Mol Pharm ; 15(9): 3625-3633, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-29771531

RESUMEN

Exosomes play a pivotal role in mediating intercellular communications and package delivery. They have recently been discovered to serve as diagnostic biomarkers as well as a possible drug delivery vehicle based on their nanometer size range and capability to transfer biological materials to recipient cells. Their unique biocompatibility, high stability, preferred tumor homing, and adjustable targeting efficiency can make exosomes an attractive and potentially effective tool of drug delivery in cancer therapy. While exosomes possess properties that make them uniquely suitable for delivery of bioactive molecules, there remains a to-be-filled gap between the current understanding about exosome biology and the ideal application scenarios. In this review, we summarize the characteristics enabling the potential of exosomes for drug delivery as well as the outstanding questions related to exosome composition and function, production and purification, bioengineering and targeting, uptake and biodistribution, efficacy and immune regulation, etc. Advanced technologies are demanded to visualize, characterize, and sort heterogeneous exosome populations. We are positive that the deeper and more comprehensive understanding of exosome biology as well as advanced nanotechnology will certainly accelerate its therapeutic applications.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Exosomas/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Bioingeniería/métodos , Comunicación Celular/fisiología , Humanos , Neoplasias/patología
19.
Anticancer Res ; 38(2): 647-654, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29374686

RESUMEN

BACKGROUND/AIM: Sirtuins (SIRTs) play crucial roles in various signaling pathways that modulate differentiation and proliferation. We sought to elucidate the role of SIRTs in differentiation and proliferation of human neuroblastoma (NB). MATERIALS AND METHODS: NB cells were treated with nicotinamide (NAM), a non-specific SIRT inhibitor, SIRT-targeted short hairpin RNAs, and retinoic acid to assess cell growth and differentiation. RESULTS: SIRTs are involved in proliferation and differentiation using NAM in BE(2)-C cells. Specifically, SIRT6 knockdown in BE(2)-C cells reduced cell proliferation, induced neurite extension, corresponding with induction of p21CIP1 expression and G1 cell-cycle arrest. These effects were rescued by forced re-overexpression of SIRT6. SIRT6 expression was reduced in differentiated human NB sections, and RA-induced differentiation in BE(2)-C cells. CONCLUSION: SIRTs have important oncogenic properties in NB beyond its established functions in aging and genome stability. SIRT6 may represent a novel target for developing future therapeutics for the treatment of aggressive NBs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Sirtuinas/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Técnicas de Silenciamiento del Gen , Humanos , Neuroblastoma/patología , Niacinamida/farmacología , ARN Interferente Pequeño/farmacología , Sirtuinas/genética , Tretinoina/administración & dosificación , Tretinoina/farmacología
20.
Clin Cancer Res ; 24(7): 1516-1517, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29358501

RESUMEN

Extracellular vesicles containing glycogen phosphorylase, brain/heart (PYGB) have been demonstrated as a sensitive biomarker for normal cardiac injuries for patients after chemotherapy. Oxidative stress was suggested to be the mechanism behind the chemotherapy-induced tissue damage and augmented with mitochondrial antioxidant could be an effective means of early intervention. Clin Cancer Res; 24(7); 1516-7. ©2018 AACRSee related article by Yarana et al., p. 1644.


Asunto(s)
Vesículas Extracelulares , Miocitos Cardíacos/efectos de los fármacos , Animales , Biomarcadores , Doxorrubicina , Glucógeno Fosforilasa , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA