Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Nanobiotechnology ; 21(1): 352, 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37770932

ABSTRACT

BACKGROUND: Macrophages are highly plastic innate immune cells that play key roles in host defense, tissue repair, and homeostasis maintenance. In response to divergent stimuli, macrophages rapidly alter their functions and manifest a wide polarization spectrum with two extremes: M1 or classical activation and M2 or alternative activation. Extracellular vesicles (EVs) secreted from differentially activated macrophages have been shown to have diverse functions, which are primarily attributed to their microRNA cargos. The role of protein cargos in these EVs remains largely unexplored. Therefore, in this study, we focused on the protein cargos in macrophage-derived EVs. RESULTS: Naïve murine bone marrow-derived macrophages were treated with lipopolysaccharide or interlukin-4 to induce M1 or M2 macrophages, respectively. The proteins of EVs and their parental macrophages were subjected to quantitative proteomics analyses, followed by bioinformatic analyses. The enriched proteins of M1-EVs were involved in proinflammatory pathways and those of M2-EVs were associated with immunomodulation and tissue remodeling. The signature proteins of EVs shared a limited subset of the proteins of their respective progenitor macrophages, but they covered many of the typical pathways and functions of their parental cells, suggesting their respective M1-like and M2-like phenotypes and functions. Experimental examination validated that protein cargos in M1- or M2-EVs induced M1 or M2 polarization, respectively. More importantly, proteins in M1-EVs promoted viability, proliferation, and activation of T lymphocytes, whereas proteins in M2-EVs potently protected the tight junction structure and barrier integrity of epithelial cells from disruption. Intravenous administration of M2-EVs in colitis mice led to their accumulation in the colon, alleviation of colonic inflammation, promotion of M2 macrophage polarization, and improvement of gut barrier functions. Protein cargos in M2-EVs played a key role in their protective function in colitis. CONCLUSION: This study has yielded a comprehensive unbiased dataset of protein cargos in macrophage-derived EVs, provided a systemic view of their potential functions, and highlighted the important engagement of protein cargos in the pathophysiological functions of these EVs.


Subject(s)
Colitis , Extracellular Vesicles , Animals , Mice , Macrophages/metabolism , Phagocytosis , Extracellular Vesicles/metabolism , Colitis/metabolism , Inflammation/metabolism
2.
Antioxidants (Basel) ; 11(4)2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35453374

ABSTRACT

Arsenic, a naturally occurring metalloid derived from the environment, has been studied worldwide for its causative effects in various cancers. However, the effects of arsenic toxicity on the development and progression of metabolic syndrome, including obesity and diabetes, has received less attention. Many studies suggest that metabolic dysfunction and autophagy dysregulation of adipose and muscle tissues are closely related to the development of metabolic disease. In the USA, arsenic contamination has been reported in some ground water, soil and grain samples in major agricultural regions, but the effects on adipose and muscle tissue metabolism and autophagy have not been investigated much. Here, we highlight arsenic toxicity according to the species, dose and exposure time and the effects on adipose and muscle tissue metabolism and autophagy. Historically, arsenic was used as both a poison and medicine, depending on the dose and treatment time. In the modern era, arsenic intoxication has significantly increased due to exposure from water, soil and food, which could be a contributing factor in the development and progression of metabolic disease. From this review, a better understanding of the pathogenic mechanisms by which arsenic alters metabolism and autophagy regulation could become a cornerstone leading to the development of therapeutic strategies against arsenic-induced toxicity and metabolic disease.

3.
Theranostics ; 11(19): 9311-9330, 2021.
Article in English | MEDLINE | ID: mdl-34646372

ABSTRACT

Aberrant activation of the nucleotide-binding domain and leucine-rich repeat related (NLR) family, pyrin domain containing 3 (NLRP3) inflammasome drives the development of many complex inflammatory diseases, such as obesity, Alzheimer's disease, and atherosclerosis. However, no medications specifically targeting the NLRP3 inflammasome have become clinically available. Therefore, we aim to identify new inhibitors of the NLRP3 inflammasome in this study. Methods: Vesicle-like nanoparticles (VLNs) were extracted from garlic chives and other Allium vegetables and their effects on the NLRP3 inflammasome were evaluated in primary macrophages. After garlic chive-derived VLNs (GC-VLNs) were found to exhibit potent anti-NLRP3 inflammasome activity in cell culture, such function was further assessed in a murine acute liver injury disease model, as well as in diet-induced obesity. Finally, GC-VLNs were subjected to omics analysis to identify the active components with anti-NLRP3 inflammasome function. Results: GC-VLNs are membrane-enclosed nanoparticles containing lipids, proteins, and RNAs. They dose-dependently inhibit pathways downstream of NLRP3 inflammasome activation, including caspase-1 autocleavage, cytokine release, and pyroptotic cell death in primary macrophages. The inhibitory effects of GC-VLNs on the NLRP3 inflammasome are specific, considering their marginal impact on activation of other inflammasomes. Local administration of GC-VLNs in mice alleviates NLRP3 inflammasome-mediated inflammation in chemical-induced acute liver injury. When administered orally or intravenously, GC-VLNs accumulate in specific tissues and suppress activation of the NLRP3 inflammasome and chronic inflammation in diet-induced obese mice. The phospholipid 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC) in GC-VLNs has been identified to inhibit NLRP3 inflammasome activation. Conclusions: Identification of GC-VLNs and their active component DLPC as potent inflammasome inhibitors provides new therapeutic candidates in the treatment of NLRP3 inflammasome-driven diseases.


Subject(s)
Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Plant Extracts/pharmacology , Animals , Antioxidants/pharmacology , China , Chive/metabolism , Cytokines/metabolism , Drug Evaluation, Preclinical/methods , Extracellular Vesicles/metabolism , Garlic/metabolism , Inflammation/metabolism , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nanoparticles/chemistry , Obesity , Phagocytosis
4.
Am J Physiol Cell Physiol ; 321(3): C607-C614, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34378992

ABSTRACT

Bovine milk exosomes (BMEs) are being explored in drug delivery despite their rapid elimination by macrophages. We aimed at identifying the BME transporter in murine bone marrow-derived macrophages (BMDMs). Fluorophore-labeled BMEs were used in transport studies in BMDMs from C57BL/6J and class A scavenger receptor type 1/2 (CASR-1/2) knockout mice and tissue accumulation in macrophage-depleted C57BL/6J mice. Parametric and nonparametric statistics tests for pairwise and multiple comparisons were used. Chemical inhibitors of phagocytosis by cytochalasin D led to a 69 ± 18% decrease in BME uptake compared with controls (P < 0.05), whereas inhibitors of endocytic pathways other than phagocytosis had a modest effect on uptake (P > 0.05). Inhibitors of class A scavenger receptors (CASRs) including CASR-1/2 caused a 70% decrease in BME uptake (P < 0.05). The uptake of BMEs by BMDMs from CASR-1/2 knockout mice was smaller by 58 ± 23% compared with wild-type controls (P < 0.05). Macrophage depletion by clodronate caused a more than 44% decrease in BME uptake in the spleen and lungs (P < 0.05), whereas the decrease observed in liver was not statistically significant. In conclusion, CASR-1/2 facilitates the uptake of BMEs in BMDMs and C57BL/6J mice.


Subject(s)
Exosomes/metabolism , Macrophages/metabolism , Milk/chemistry , Scavenger Receptors, Class A/genetics , Animals , Cattle , Clodronic Acid/pharmacology , Cytochalasin D/pharmacology , Endocytosis/drug effects , Exosomes/chemistry , Female , Fluorescent Dyes/chemistry , Gene Expression , Liver/drug effects , Liver/metabolism , Lung/drug effects , Lung/metabolism , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Protein Isoforms/deficiency , Protein Isoforms/genetics , Scavenger Receptors, Class A/deficiency , Spleen/drug effects , Spleen/metabolism , Staining and Labeling/methods
5.
Biomedicines ; 9(2)2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33535473

ABSTRACT

The nucleotide-binding domain and leucine-rich repeat related (NLR) family, pyrin domain containing 3 (NLRP3) inflammasome is a multimeric protein complex that recognizes various danger or stress signals from pathogens, the host, and the environment, leading to activation of caspase-1 and inducing inflammatory responses. This pro-inflammatory protein complex plays critical roles in pathogenesis of a wide range of diseases including neurodegenerative diseases, autoinflammatory diseases, and metabolic disorders. Therefore, intensive efforts have been devoted to understanding its activation mechanisms and to searching for its specific inhibitors. Approximately forty natural compounds with anti-NLRP3 inflammasome properties have been identified. Here, we provide an update about new natural compounds that have been identified within the last three years to inhibit the NLRP3 inflammasome and offer an overview of the underlying molecular mechanisms of their anti-NLRP3 inflammasome activities.

6.
J Extracell Vesicles ; 10(4): e12069, 2021 02.
Article in English | MEDLINE | ID: mdl-33613874

ABSTRACT

Honey has been used as a nutrient, an ointment, and a medicine worldwide for many centuries. Modern research has demonstrated that honey has many medicinal properties, reflected in its anti-microbial, anti-oxidant, and anti-inflammatory bioactivities. Honey is composed of sugars, water and a myriad of minor components, including minerals, vitamins, proteins and polyphenols. Here, we report a new bioactive component‒vesicle-like nanoparticles‒in honey (H-VLNs). These H-VLNs are membrane-bound nano-scale particles that contain lipids, proteins and small-sized RNAs. The presence of plant-originated plasma transmembrane proteins and plasma membrane-associated proteins suggests the potential vesicle-like nature of these particles. H-VLNs impede the formation and activation of the nucleotide-binding domain and leucine-rich repeat related (NLR) family, pyrin domain containing 3 (NLRP3) inflammasome, which is a crucial inflammatory signalling platform in the innate immune system. Intraperitoneal administration of H-VLNs in mice alleviates inflammation and liver damage in the experimentally induced acute liver injury. miR-4057 in H-VLNs was identified in inhibiting NLRP3 inflammasome activation. Together, our studies have identified anti-inflammatory VLNs as a new bioactive agent in honey.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Honey/analysis , Inflammasomes/metabolism , Inflammation/metabolism , MicroRNAs/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nanoparticles/chemistry , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Bees/metabolism , Cells, Cultured , Disease Models, Animal , Extracellular Vesicles/chemistry , Immunity, Innate , Insect Proteins/analysis , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Nanoparticles/ultrastructure , Plant Proteins/analysis , Proteomics , Signal Transduction
7.
Nutrients ; 12(2)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32069862

ABSTRACT

Fulminant hepatic failure (FHF) is a rare, life-threatening liver disease with a poor prognosis. Administration of D-galactosamine (GalN) and lipopolysaccharide (LPS) triggers acute liver injury in mice, simulating many clinical features of FHF in humans; therefore, this disease model is often used to investigate potential therapeutic interventions to treat FHF. Recently, suppression of the nucleotide-binding domain and leucine-rich repeat related (NLR) family, pyrin domain containing 3 (NLRP3) inflammasome, was shown to alleviate the severity of GalN/LPS-induced liver damage in mice. Therefore, the goal of this study was to find dietary exosome-like nanoparticles (ELNs) with therapeutic potential in curbing FHF by suppressing the NLRP3 inflammasome. Seven commonly consumed mushrooms were used to extract ELNs. These mushrooms were found to contain ELNs composed of RNAs, proteins, and lipids. Among these mushroom-derived ELNs, only shiitake mushroom-derived ELNs (S-ELNs) substantially inhibited NLRP3 inflammasome activation by preventing inflammasome formation in primary macrophages. S-ELNs also suppressed the secretion of interleukin (IL)-6, as well as both protein and mRNA levels of the Il1b gene. Remarkably, pre-treatment with S-ELNs protected mice from GalN/LPS-induced acute liver injury. Therefore, S-ELNs, identified as potent new inhibitors of the NLRP3 inflammasome, represent a promising class of agents with the potential to combat FHF.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Nanoparticles/chemistry , Plant Extracts/pharmacology , Protective Agents/pharmacology , Shiitake Mushrooms/chemistry , Animals , Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Exosomes , Galactosamine , Lipopolysaccharides , Liver/metabolism , Liver Failure, Acute/drug therapy , Liver Failure, Acute/etiology , Macrophages/drug effects , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects
8.
Mol Pharm ; 16(6): 2690-2699, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31038962

ABSTRACT

The nucleotide-binding domain and leucine-rich repeat-containing family, pyrin domain-containing 3 (NLRP3) inflammasome is a key regulator of innate immune responses, and its aberrant activation is implicated in the pathogenesis of many diseases such as Alzheimer's disease and type 2 diabetes. Targeting the NLRP3 inflammasome could hold promise to combat these complex diseases, but therapies specifically inhibiting the NLRP3 inflammasome have not been developed for patient treatment. The current study aimed to identify food-borne exosome-like nanoparticles (ELNs) that inhibit NLRP3 inflammasome activity. Nine vegetables or fruits were selected to extract ELNs, which were examined for their inhibitory effects on activation of the NLRP3 inflammasome in primary macrophages. Although most of the tested ELNs posed minimal impacts, the ELNs from ginger rhizomes (G-ELNs) strongly inhibited NLRP3 inflammasome activation. The G-ELNs contained lipids, proteins, and RNAs and were easily taken up by macrophages. G-ELN treatment suppressed pathways downstream of inflammasome activation including caspase1 autocleavage, interleukin (IL)-1ß and IL-18 secretion, and pyroptotic cell death. Apoptotic speck protein containing a caspase recruitment domain (ASC) oligomerization and speck formation assays indicated that G-ELNs blocked assembly of the NLRP3 inflammasome. The lipids in G-ELNs, rather than the RNAs or proteins, were responsible for the inhibitory activity observed. Together, the data suggested G-ELNs as new potent agents that block NLRP3 inflammasome assembly and activation. The unique features of G-ELNs including biomolecule protection and tissue bioavailability should facilitate the development of G-ELN-based therapy to target the NLRP3 inflammasome in the disease settings.


Subject(s)
Exosomes/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nanoparticles/chemistry , Rhizome/metabolism , Zingiber officinale , Animals , Chromatography, Thin Layer , Immunoblotting , Inflammasomes/metabolism , Liposomes/metabolism , Mice , Mice, Inbred C57BL
9.
Nat Commun ; 9(1): 960, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29511190

ABSTRACT

Majority of disease-modifying therapeutic targets are restricted to the intracellular space and are therefore not druggable using existing biologic modalities. The ability to efficiently deliver macromolecules inside target cells or tissues would greatly expand the current landscape of therapeutic targets for future generations of biologic drugs, but remains challenging. Here we report the use of extracellular vesicles, known as arrestin domain containing protein 1 [ARRDC1]-mediated microvesicles (ARMMs), for packaging and intracellular delivery of a myriad of macromolecules, including the tumor suppressor p53 protein, RNAs, and the genome-editing CRISPR-Cas9/guide RNA complex. We demonstrate selective recruitment of these macromolecules into ARMMs. When delivered intracellularly via ARMMs, these macromolecules are biologically active in recipient cells. P53 delivered via ARMMs induces DNA damage-dependent apoptosis in multiple tissues in mice. Together, our results provide proof-of-principle demonstration that ARMMs represent a highly versatile platform for packaging and intracellular delivery of therapeutic macromolecules.


Subject(s)
Arrestins/metabolism , Drug Carriers/metabolism , Extracellular Vesicles/metabolism , A549 Cells , Animals , CRISPR-Associated Protein 9/administration & dosage , Green Fluorescent Proteins , HEK293 Cells , Humans , Mice, Knockout , RNA/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Tumor Suppressor Protein p53/administration & dosage , Tumor Suppressor Protein p53/genetics
10.
Elife ; 52016 Feb 19.
Article in English | MEDLINE | ID: mdl-26894960

ABSTRACT

Macrophage activation/polarization to distinct functional states is critically supported by metabolic shifts. How polarizing signals coordinate metabolic and functional reprogramming, and the potential implications for control of macrophage activation, remains poorly understood. Here we show that IL-4 signaling co-opts the Akt-mTORC1 pathway to regulate Acly, a key enzyme in Ac-CoA synthesis, leading to increased histone acetylation and M2 gene induction. Only a subset of M2 genes is controlled in this way, including those regulating cellular proliferation and chemokine production. Moreover, metabolic signals impinge on the Akt-mTORC1 axis for such control of M2 activation. We propose that Akt-mTORC1 signaling calibrates metabolic state to energetically demanding aspects of M2 activation, which may define a new role for metabolism in supporting macrophage activation.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Macrophage Activation , Macrophages/metabolism , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Acetylation , Animals , Cell Proliferation , Chemokines/metabolism , Gene Expression Regulation , Histones/metabolism , Interleukin-4/metabolism , Macrophages/physiology , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Protein Processing, Post-Translational
11.
Proc Natl Acad Sci U S A ; 111(43): 15514-9, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25313054

ABSTRACT

Inflammasomes are intracellular sensors that couple detection of pathogens and cellular stress to activation of Caspase-1, and consequent IL-1ß and IL-18 maturation and pyroptotic cell death. Here, we show that the absent in melanoma 2 (AIM2) and nucleotide-binding oligomerization domain-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasomes trigger Caspase-1-dependent mitochondrial damage. Caspase-1 activates multiple pathways to precipitate mitochondrial disassembly, resulting in mitochondrial reactive oxygen species (ROS) production, dissipation of mitochondrial membrane potential, mitochondrial permeabilization, and fragmentation of the mitochondrial network. Moreover, Caspase-1 inhibits mitophagy to amplify mitochondrial damage, mediated in part by cleavage of the key mitophagy regulator Parkin. In the absence of Parkin activity, increased mitochondrial damage augments pyroptosis, as indicated by enhanced plasma membrane permeabilization and release of danger-associated molecular patterns (DAMPs). Therefore, like other initiator caspases, Caspase-1 activation by inflammasomes results in mitochondrial damage.


Subject(s)
Caspase 1/metabolism , Inflammasomes/metabolism , Mitochondria/pathology , Mitophagy , Animals , Apoptosis , Carrier Proteins/metabolism , Cryopyrin-Associated Periodic Syndromes/enzymology , Cryopyrin-Associated Periodic Syndromes/pathology , DNA-Binding Proteins/metabolism , Humans , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Permeability , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
12.
Sci Rep ; 3: 2806, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24076663

ABSTRACT

Sirt5, localized in the mitochondria, is a member of sirtuin family of NAD⁺-dependent deacetylases. Sirt5 was shown to deacetylate and activate carbamoyl phosphate synthase 1. Most recently, Sirt5 was reported to be the predominant protein desuccinylase and demalonylase in the mitochondria because the ablation of Sirt5 enhanced the global succinylation and malonylation of mitochondrial proteins, including many metabolic enzymes. In order to determine the physiological role of Sirt5 in metabolic homeostasis, we generated a germline Sirt5 deficient (Sirt5⁻/⁻) mouse model and performed a thorough metabolic characterization of this mouse line. Although a global protein hypersuccinylation and elevated serum ammonia during fasting were observed in our Sirt5⁻/⁻ mouse model, Sirt5 deficiency did not lead to any overt metabolic abnormalities under either chow or high fat diet conditions. These observations suggest that Sirt5 is likely to be dispensable for the metabolic homeostasis under the basal conditions.


Subject(s)
Metabolome , Sirtuins/deficiency , Animals , Diet, High-Fat , Feeding Behavior , Male , Metabolome/genetics , Mice , Mice, Inbred C57BL , Models, Animal , Phenotype , Reproducibility of Results , Sirtuins/metabolism , Transcription, Genetic
13.
Proc Natl Acad Sci U S A ; 109(28): 11282-7, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733741

ABSTRACT

The NLRP3 (nucleotide-binding domain, leucine-rich-repeat-containing family, pyrin domain-containing 3) inflammasome mediates production of inflammatory mediators, such as IL-1ß and IL-18, and as such is implicated in a variety of inflammatory processes, including infection, sepsis, autoinflammatory diseases, and metabolic diseases. The proximal steps in NLRP3 inflammasome activation are not well understood. Here we elucidate a critical role for Ca(2+) mobilization in activation of the NLRP3 inflammasome by multiple stimuli. We demonstrate that blocking Ca(2+) mobilization inhibits assembly and activation of the NLRP3 inflammasome complex, and that during ATP stimulation Ca(2+) signaling is pivotal in promoting mitochondrial damage. C/EPB homologous protein, a transcription factor that can modulate Ca(2+) release from the endoplasmic reticulum, amplifies NLRP3 inflammasome activation, thus linking endoplasmic reticulum stress to activation of the NLRP3 inflammasome. Our findings support a model for NLRP3 inflammasome activation by Ca(2+)-mediated mitochondrial damage.


Subject(s)
Calcium/metabolism , Carrier Proteins/metabolism , Inflammasomes/metabolism , Adenosine Triphosphate/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Flow Cytometry/methods , Immunity, Innate , Inflammation/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Models, Biological , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction
14.
EMBO Rep ; 12(10): 1069-76, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21836635

ABSTRACT

The nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase SIRT1 is a major metabolic regulator activated by energy stresses such as fasting or calorie restriction. SIRT1 activation during fasting not only relies on the increase in the NAD(+)/NADH ratio caused by energy deprivation but also involves an upregulation of SIRT1 mRNA and protein levels in various metabolic tissues. We demonstrate that SIRT1 expression is controlled systemically by the activation of the cyclic AMP response-element-binding protein upon low nutrient availability. Conversely, in the absence of energetic stress, the carbohydrate response-element-binding protein represses the expression of SIRT1. Altogether, these results demonstrate that SIRT1 expression is tightly controlled at the transcriptional level by nutrient availability and further underscore that SIRT1 is a crucial metabolic checkpoint connecting the energetic status with transcriptional programmes.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Sirtuin 1/genetics , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , CHO Cells , Cell Line, Tumor , Cricetinae , Fasting , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Sirtuin 1/metabolism , Transcriptional Activation
15.
Pharmacol Res ; 62(1): 35-41, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20026274

ABSTRACT

The biological function of most proteins relies on reversible post-translational modifications, among which phosphorylation is most prominently studied and well recognized. Recently, a growing amount of evidence indicates that acetylation-deacetylation reactions, when applied to crucial mediators, can also robustly affect the function of target proteins and thereby have wide-ranging physiological impacts. Sirtuin 1 (SIRT1), which functions as a nicotinamide adenine dinucleotide (NAD(+))-dependent protein deacetylase, deacetylates a wide variety of metabolic molecules in response to the cellular energy and redox status and as such causes significant changes in metabolic homeostasis. This review surveys the evidence for the emerging role of SIRT1-mediated deacetylation in the control of metabolic homeostasis.


Subject(s)
Energy Metabolism/physiology , Protein Processing, Post-Translational , Sirtuin 1/physiology , Acetylation , Animals , Humans , Mitochondria/metabolism , Oxidation-Reduction , Sirtuin 1/metabolism , Transcription Factors/metabolism
16.
Ann N Y Acad Sci ; 1173 Suppl 1: E10-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19751409

ABSTRACT

Recently the function of the sirtuin family, named after their homology to the Saccharomyces cerevisiae gene silent information regulator 2 (Sir2), has received a lot of attention, as their beneficial impact on longevity was linked to their effects on metabolic control. All sirtuins require nicotinamide adenine dinucleotide (NAD(+)) for their deacetylase or ADP-ribosyl transferase activity, linking their function tightly to cellular energy levels. SIRT1, the founding member of the sirtuin family, modulates many aspects of glucose and lipid homeostasis in almost all key metabolic tissues. Other members including SIRT2, SIRT3, and SIRT4 are also implicated in various metabolic processes. Here, we review the recent data related to the role of sirtuins in the control of metabolic homeostasis and possible underlying molecular mechanisms.


Subject(s)
Homeostasis/physiology , Longevity/physiology , Sirtuins/physiology , Adipose Tissue/physiology , Energy Metabolism , Glucose/metabolism , Humans , Insulin/physiology , Lipids/physiology , Mitochondria/physiology , Muscle, Skeletal/physiology , NAD/metabolism , Sirtuin 1 , Sirtuin 2 , Sirtuins/genetics , Sirtuins/metabolism , Thermogenesis
17.
Cancer Res ; 66(18): 9316-22, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16982777

ABSTRACT

Cells are frequently challenged by DNA double-strand breaks (DSB) that threaten their normal function and survival. In mammalian cells, the repair of DSBs is predominantly mediated by the DNA-dependent protein kinase (DNA-PK) complex. We unexpectedly found that the corepressor silencing mediator for retinoid and thyroid hormone receptor (SMRT) associates with the DNA-PK repair complex. The SMRT/histone deacetylase 3 complex is required for the transcriptional repressive property of the Ku70 subunit of the repair complex. Moreover, SMRT, but not the related Nuclear Receptor Corepressor, is required for cellular recovery from DNA DSBs induced by ionizing radiation or DNA damage-inducing drugs. Thus, the corepressor SMRT plays a novel and critical role in the cellular response to DSBs.


Subject(s)
DNA Repair/physiology , DNA-Binding Proteins/physiology , Repressor Proteins/physiology , Antigens, Nuclear/metabolism , DNA Damage , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Histone Deacetylases/metabolism , Humans , Ku Autoantigen , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Substrate Specificity , Transcriptional Activation , Transfection
18.
EMBO J ; 25(17): 3966-74, 2006 Sep 06.
Article in English | MEDLINE | ID: mdl-16917504

ABSTRACT

Unliganded thyroid hormone receptor (TR) actively represses transcription via the nuclear receptor corepressor (N-CoR)/histone deacetylase 3 (HDAC3) complex. Although transcriptional activation by liganded receptors involves chromatin remodeling, the role of ATP-dependent remodeling in receptor-mediated repression is unknown. Here we report that SNF2H, the mammalian ISWI chromatin remodeling ATPase, is critical for repression of a genomically integrated, TR-regulated reporter gene. N-CoR and HDAC3 are both required for recruitment of SNF2H to the repressed gene. SNF2H does not interact directly with the N-CoR/HDAC3 complex, but binds to unacetylated histone H4 tails, suggesting that deacetylase activity of the corepressor complex is critical to SNF2H function. Indeed, HDAC3 as well as SNF2H are required for nucleosomal organization on the TR target gene. Consistent with these findings, reduction of SNF2H induces expression of an endogenous TR-regulated gene, dio1, in liver cells. Thus, although not apparent from studies of transiently transfected reporter genes, gene repression by TR involves the targeting of chromatin remodeling factors to repressed genes by the HDAC activity of nuclear receptor corepressors.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Nuclear Proteins/metabolism , Receptors, Thyroid Hormone/physiology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Adenosine Triphosphatases/genetics , Animals , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Genes, Reporter , Histone Deacetylases/genetics , Histones/metabolism , Humans , Luciferases/genetics , Mice , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Promoter Regions, Genetic , RNA Interference , Repressor Proteins/genetics , Transcription Factors/genetics
19.
EMBO Rep ; 6(5): 445-51, 2005 May.
Article in English | MEDLINE | ID: mdl-15832170

ABSTRACT

Ligands for nuclear receptors facilitate the exchange of co-repressors for coactivators, leading to chromatin modifications that favour the activation of gene transcription. Here, we show that the repressed state of an endogenous retinoic acid-regulated gene is quickly re-established after ligand removal. As expected, repression is characterized by recruitment of N-CoR/SMRT-HDAC3 (histone deacetylase 3) co-repressor complexes, leading to local histone hypoacetylation. The achievement of the repressed state involves the ordered deacetylation of lysines in H4 tails. This order is determined by the inherent substrate specificity of HDAC3, and unexpectedly predicts the binding preference of N-CoR/SMRT for submaximally acetylated H4 tails. The match between the specificity of acetyl-histone deacetylation by HDAC3 and the histone-binding preference of N-CoR/SMRT allows the co-repressor complex to stabilize and propagate repression of nuclear hormone receptor gene targets.


Subject(s)
Histone Code , Histone Deacetylases/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , 3T3-L1 Cells , Acetylation , Animals , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histones/metabolism , Ligands , Mice , Mucoproteins/metabolism , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Protein Binding , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Substrate Specificity , Tretinoin/pharmacology , Retinoic Acid Receptor gamma
20.
J Biol Chem ; 280(8): 7346-58, 2005 Feb 25.
Article in English | MEDLINE | ID: mdl-15561719

ABSTRACT

The identities of signal transducer proteins that integrate histone hypoacetylation and transcriptional repression are largely unknown. Here we demonstrate that THAP7, an uncharacterized member of the recently identified THAP (Thanatos-associated protein) family of proteins, is ubiquitously expressed, associates with chromatin, and represses transcription. THAP7 binds preferentially to hypoacetylated (un-, mono-, and diacetylated) histone H4 tails in vitro via its C-terminal 77 amino acids. Deletion of this domain, or treatment of cells with the histone deacetylase inhibitor TSA, which leads to histone hyperacetylation, partially disrupts THAP7/chromatin association in living cells. THAP7 coimmunoprecipitates with histone deacetylase 3 (HDAC3) and the nuclear hormone receptor corepressor (NCoR) and represses transcription as a Gal4 fusion protein. Chromatin immunoprecipitation assays demonstrate that these corepressors are recruited to promoters in a THAP7 dependent manner and promote histone H3 hypoacetylation. The conserved THAP domain is a key determinant for full HDAC3 association in vitro, and both the THAP domain and the histone interaction domain are important for the repressive properties of THAP7. Full repression mediated by THAP7 is also dependent on NCoR expression. We hypothesize that THAP7 is a dual function repressor protein that actively targets deacetylation of histone H3 necessary to establish transcriptional repression and functions as a signal transducer of the repressive mark of hypoacetylated histone H4. This is the first demonstration of the transcriptional regulatory properties of a human THAP domain protein, and a critical identification of a potential transducer of the repressive signal of hypoacetylated histone H4 in higher eukaryotes.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins/physiology , Histone Deacetylases/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Repressor Proteins/metabolism , Repressor Proteins/physiology , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Protein Binding , Protein Transport , Repressor Proteins/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...