Browsing by Subject "Acetylation"
Results Per Page
Sort Options
Item Open Access Broad Remodeling of the Acetylproteome by SIRT3 Manipulation Fails to Affect Insulin Secretion or β-cell Metabolism in the Absence of Dietary Overnutrition(2018) Peterson, Brett StevenSIRT3 is an NAD+-dependent mitochondrial protein deacetylase purported to influence cellular and systemic metabolism through modulation of the mitochondrial acetylproteome. Fuel-stimulated insulin secretion from pancreatic islets involves mitochondrial metabolism and might be susceptible to SIRT3-mediated effects. To investigate this idea, we used CRISPR/Cas9 technology to obtain complete SIRT3 knockout in the INS-1 832/13 insulinoma cell line. In the context of this SIRT3 knockout cell line, we re-expressed wild-type SIRT3, β-Galactosidase, or one of three enzymatically inactive mutant forms of SIRT3 to generate lines representing a wide range of SIRT3 expression and mitochondrial protein deacetylase activity. We performed large-scale acetylproteome profiling by mass spectrometry on the different lines, and observed wide-spread, SIRT3-dependent changes in acetylation of enzymes involved in fatty acid oxidation, the TCA cycle, and the electron transport chain. Remarkably, despite these broad changes, the cell lines had indistinguishable insulin secretion responses to glucose or pyruvate, and exhibited no differences in function or viability in response to metabolic or ER stress-inducing agents. Moreover, metabolomic profiling revealed that, when compared to SIRT3-null cell lines, expression of wild-type SIRT3 does not result in appreciable changes in a host of organic acid, amino acid or fatty acid-derived acylcarnitine metabolites during glucose stimulation.
We also studied mice with global SIRT3 knockout (KO) fed a standard chow (STD) or high-fat/high-sucrose (HFHS) diet. Importantly, we performed these studies in the C57Bl/6J background in which we replaced the mutant allele of nicotinamide nucleotide transhydrogenase (NNT) present in the “J” substrain, with the wild-type allele in order to restore endogenous NNT function. SIRT3 KO and wild-type (WT) mice fed a STD diet exhibited no differences in insulin secretion during oral or IP glucose tolerance tests, and the function of islets isolated from these mice was indistinguishable in islet perifusion studies conducted with a broad array of secretagogues. Only when chronically fed a HFHS diet did SIRT3 KO animals exhibit a modest impairment in insulin secretion, but without an effect on glycemic control. Our broad conclusion is that major changes in mitochondrial protein acetylation in response to manipulation of SIRT3 are not sufficient to cause changes in islet function or metabolism. However, under conditions of chronic nutritional stress (feeding of a HFHS diet for 12 weeks), a negative effect on function appears, suggesting that islets are more susceptible to nutrition-induced factors (oxidative stress, local cytokine production, etc.) when SIRT3 is absent. Further studies will be required to identify factors that may interact with SIRT3 deficiency and mitochondrial protein hyperacetylation to increase the risk of -cell dysfunction.
Item Open Access Cigarette smoke modulates vascular smooth muscle phenotype: implications for carotid and cerebrovascular disease.(PloS one, 2013-01) Starke, Robert M; Ali, Muhammad S; Jabbour, Pascal M; Tjoumakaris, Stavropoula I; Gonzalez, Fernando; Hasan, David M; Rosenwasser, Robert H; Owens, Gary K; Koch, Walter J; Dumont, Aaron SBackground
The role of smooth muscle cell (SMC) phenotypic modulation in the cerebral circulation and pathogenesis of stroke has not been determined. Cigarette smoke is a major risk factor for atherosclerosis, but potential mechanisms are unclear, and its role in SMC phenotypic modulation has not been established.Methods and results
In cultured cerebral vascular SMCs, exposure to cigarette smoke extract (CSE) resulted in decreased promoter activity and mRNA expression of key SMC contractile genes (SM-α-actin, SM-22α, SM-MHC) and the transcription factor myocardin in a dose-dependent manner. CSE also induced pro-inflammatory/matrix remodeling genes (MCP-1, MMPs, TNF-α, IL-1β, NF-κB). CSE increased expression of KLF4, a known regulator of SMC differentiation, and siKLF4 inhibited CSE induced suppression of SMC contractile genes and myocardin and activation of inflammatory genes. These mechanisms were confirmed in vivo following exposure of rat carotid arteries to CSE. Chromatin immune-precipitation assays in vivo and in vitro demonstrated that CSE promotes epigenetic changes with binding of KLF4 to the promoter regions of myocardin and SMC marker genes and alterations in promoter acetylation and methylation.Conclusion
CSE exposure results in phenotypic modulation of cerebral SMC through myocardin and KLF4 dependent mechanisms. These results provides a mechanism by which cigarette smoke induces a pro-inflammatory/matrix remodeling phenotype in SMC and an important pathway for cigarette smoke to contribute to atherosclerosis and stroke.Item Open Access CoA synthase regulates mitotic fidelity via CBP-mediated acetylation.(Nature communications, 2018-03-12) Lin, Chao-Chieh; Kitagawa, Mayumi; Tang, Xiaohu; Hou, Ming-Hsin; Wu, Jianli; Qu, Dan Chen; Srinivas, Vinayaka; Liu, Xiaojing; Thompson, J Will; Mathey-Prevot, Bernard; Yao, Tso-Pang; Lee, Sang Hyun; Chi, Jen-TsanThe temporal activation of kinases and timely ubiquitin-mediated degradation is central to faithful mitosis. Here we present evidence that acetylation controlled by Coenzyme A synthase (COASY) and acetyltransferase CBP constitutes a novel mechanism that ensures faithful mitosis. We found that COASY knockdown triggers prolonged mitosis and multinucleation. Acetylome analysis reveals that COASY inactivation leads to hyper-acetylation of proteins associated with mitosis, including CBP and an Aurora A kinase activator, TPX2. During early mitosis, a transient CBP-mediated TPX2 acetylation is associated with TPX2 accumulation and Aurora A activation. The recruitment of COASY inhibits CBP-mediated TPX2 acetylation, promoting TPX2 degradation for mitotic exit. Consistently, we detected a stage-specific COASY-CBP-TPX2 association during mitosis. Remarkably, pharmacological and genetic inactivation of CBP effectively rescued the mitotic defects caused by COASY knockdown. Together, our findings uncover a novel mitotic regulation wherein COASY and CBP coordinate an acetylation network to enforce productive mitosis.Item Open Access Core and region-enriched networks of behaviorally regulated genes and the singing genome.(Science, 2014-12-12) Whitney, Osceola; Pfenning, Andreas R; Howard, Jason T; Blatti, Charles A; Liu, Fang; Ward, James M; Wang, Rui; Audet, Jean-Nicoles; Kellis, Manolis; Mukherjee, Sayan; Sinha, Saurabh; Hartemink, Alexander J; West, Anne E; Jarvis, Erich DSongbirds represent an important model organism for elucidating molecular mechanisms that link genes with complex behaviors, in part because they have discrete vocal learning circuits that have parallels with those that mediate human speech. We found that ~10% of the genes in the avian genome were regulated by singing, and we found a striking regional diversity of both basal and singing-induced programs in the four key song nuclei of the zebra finch, a vocal learning songbird. The region-enriched patterns were a result of distinct combinations of region-enriched transcription factors (TFs), their binding motifs, and presinging acetylation of histone 3 at lysine 27 (H3K27ac) enhancer activity in the regulatory regions of the associated genes. RNA interference manipulations validated the role of the calcium-response transcription factor (CaRF) in regulating genes preferentially expressed in specific song nuclei in response to singing. Thus, differential combinatorial binding of a small group of activity-regulated TFs and predefined epigenetic enhancer activity influences the anatomical diversity of behaviorally regulated gene networks.Item Open Access Exploring the Role of Mitochondrial Bioenergetics and Metabolism in Heart Failure(2020) Davidson, Michael ThomasHeart failure is a worldwide public health problem with substantial clinical burden and economic costs. In the progression into failure, the heart undergoes dramatic alterations in mitochondrial fuel metabolism and bioenergetics. As such, there is considerable interest in the delineation of regulatory events involved in the metabolic dysfunction of heart failure. Previous collaborative work identified three metabolic signatures associated with early stage heart failure: 1) accumulation of acylcarnitine metabolites; 2) mitochondrial hyperacetylation; and 3) elevated ketone catabolism. The goal of this dissertation was to explore the role of these metabolic signatures in the pathogenesis of heart failure.
Tissue accumulation of acylcarnitine metabolites is characteristic of mitochondrial dysfunction and indicative of incomplete β-oxidation. This occurs when a large portion of the fatty acids (i.e., acyl groups) within the mitochondria are not fully catabolized and the resulting intermediates are transferred to carnitine esters, enabling the traversal of biological membranes and departure from the mitochondrial matrix.
Nϵ-acetylation in the mitochondrial matrix is a non-enzymatic, post-translational modification (PTM) that spontaneously arises from the relatively basic pH and abundance of acetyl-CoA. Accumulation of this PTM has been observed in other tissues and disease states with evidence suggesting it impairs mitochondrial metabolism and causes dysfunction. However, convincing studies are lacking to establish a direct causal connection between dysfunction and acetylation. To address this shortcoming, a novel assay platform for the comprehensive assessment of mitochondrial bioenergetic transduction was developed and validated. Next, we generated and validated a novel mouse model of cardiac mitochondrial hyperacetylation and utilized the bioenergetic assay platform to test the hypothesis that it causes metabolic perturbations. Surprisingly, these hyperacetylated mitochondria exhibited almost no deficits in mitochondrial oxidative metabolism. To determine if hyperacetylation causes mitochondrial dysfunction in vivo under pathologic stimuli, the mouse model and littermate controls were subjected to transaortic constriction, a surgical method to induce pressure-overload heart failure. The hyperacetylated animals did not exhibit enhanced sensitivity toward cardiac dysfunction relative controls. With these results, we concluded that mitochondrial hyperacetylation does not contribute to the pathogenesis of heart failure.
Elevated ketone catabolism was observed in early stage failing hearts. Through a series of murine and canine heart failure models, ketone catabolism was shown to be adaptive in response to pathological stress. Additionally, the mitochondrial bioenergetic assay platform was applied to cardiac mitochondria under substrate limited-conditions. These results indicate that ketone catabolism improves bioenergetic efficiency under constraints which mimic the failing heart. With these results, we conclude ketone catabolism is an important metabolic defense in response to the dysfunction of the failing heart.
Item Open Access HDAC inhibitors cause site-specific chromatin remodeling at PU.1-bound enhancers in K562 cells.(Epigenetics Chromatin, 2016) Frank, Christopher L; Manandhar, Dinesh; Gordân, Raluca; Crawford, Gregory EBACKGROUND: Small molecule inhibitors of histone deacetylases (HDACi) hold promise as anticancer agents for particular malignancies. However, clinical use is often confounded by toxicity, perhaps due to indiscriminate hyperacetylation of cellular proteins. Therefore, elucidating the mechanisms by which HDACi trigger differentiation, cell cycle arrest, or apoptosis of cancer cells could inform development of more targeted therapies. We used the myelogenous leukemia line K562 as a model of HDACi-induced differentiation to investigate chromatin accessibility (DNase-seq) and expression (RNA-seq) changes associated with this process. RESULTS: We identified several thousand specific regulatory elements [~10 % of total DNase I-hypersensitive (DHS) sites] that become significantly more or less accessible with sodium butyrate or suberanilohydroxamic acid treatment. Most of the differential DHS sites display hallmarks of enhancers, including being enriched for non-promoter regions, associating with nearby gene expression changes, and increasing luciferase reporter expression in K562 cells. Differential DHS sites were enriched for key hematopoietic lineage transcription factor motifs, including SPI1 (PU.1), a known pioneer factor. We found PU.1 increases binding at opened DHS sites with HDACi treatment by ChIP-seq, but PU.1 knockdown by shRNA fails to block the chromatin accessibility and expression changes. A machine-learning approach indicates H3K27me3 initially marks PU.1-bound sites that open with HDACi treatment, suggesting these sites are epigenetically poised. CONCLUSIONS: We find HDACi treatment of K562 cells results in site-specific chromatin remodeling at epigenetically poised regulatory elements. PU.1 shows evidence of a pioneer role in this process by marking poised enhancers but is not required for transcriptional activation.Item Open Access HLA-B-associated transcript 3 (Bat3)/Scythe is essential for p300-mediated acetylation of p53.(Genes Dev, 2007-04-01) Sasaki, Toru; Gan, Eugene C; Wakeham, Andrew; Kornbluth, Sally; Mak, Tak W; Okada, HitoshiIn response to DNA damage, p53 undergoes post-translational modifications (including acetylation) that are critical for its transcriptional activity. However, the mechanism by which p53 acetylation is regulated is still unclear. Here, we describe an essential role for HLA-B-associated transcript 3 (Bat3)/Scythe in controlling the acetylation of p53 required for DNA damage responses. Depletion of Bat3 from human and mouse cells markedly impairs p53-mediated transactivation of its target genes Puma and p21. Although DNA damage-induced phosphorylation, stabilization, and nuclear accumulation of p53 are not significantly affected by Bat3 depletion, p53 acetylation is almost completely abolished. Bat3 forms a complex with p300, and an increased amount of Bat3 enhances the recruitment of p53 to p300 and facilitates subsequent p53 acetylation. In contrast, Bat3-depleted cells show reduced p53-p300 complex formation and decreased p53 acetylation. Furthermore, consistent with our in vitro findings, thymocytes from Bat3-deficient mice exhibit reduced induction of puma and p21, and are resistant to DNA damage-induced apoptosis in vivo. Our data indicate that Bat3 is a novel and essential regulator of p53-mediated responses to genotoxic stress, and that Bat3 controls DNA damage-induced acetylation of p53.Item Open Access The bromodomain protein Brd4 insulates chromatin from DNA damage signalling.(Nature, 2013-06-13) Floyd, Scott R; Pacold, Michael E; Huang, Qiuying; Clarke, Scott M; Lam, Fred C; Cannell, Ian G; Bryson, Bryan D; Rameseder, Jonathan; Lee, Michael J; Blake, Emily J; Fydrych, Anna; Ho, Richard; Greenberger, Benjamin A; Chen, Grace C; Maffa, Amanda; Del Rosario, Amanda M; Root, David E; Carpenter, Anne E; Hahn, William C; Sabatini, David M; Chen, Clark C; White, Forest M; Bradner, James E; Yaffe, Michael BDNA damage activates a signalling network that blocks cell-cycle progression, recruits DNA repair factors and/or triggers senescence or programmed cell death. Alterations in chromatin structure are implicated in the initiation and propagation of the DNA damage response. Here we further investigate the role of chromatin structure in the DNA damage response by monitoring ionizing-radiation-induced signalling and response events with a high-content multiplex RNA-mediated interference screen of chromatin-modifying and -interacting genes. We discover that an isoform of Brd4, a bromodomain and extra-terminal (BET) family member, functions as an endogenous inhibitor of DNA damage response signalling by recruiting the condensin II chromatin remodelling complex to acetylated histones through bromodomain interactions. Loss of this isoform results in relaxed chromatin structure, rapid cell-cycle checkpoint recovery and enhanced survival after irradiation, whereas functional gain of this isoform compacted chromatin, attenuated DNA damage response signalling and enhanced radiation-induced lethality. These data implicate Brd4, previously known for its role in transcriptional control, as an insulator of chromatin that can modulate the signalling response to DNA damage.Item Open Access The Role of Acetylation in the Metabolic Reprogramming of Cancer Cells(2016) McDonnell, EoinIdentifying metabolic vulnerabilities of cancer cells remains a subject of investigation for the identification of potential metabolically based therapies for cancer. It is well known that proliferating cells become largely dependent on glucose and glutamine for their growth. Interestingly, we find that lipid oxidizing genes are consistently downregulated across a wide variety of cancers while lipid synthesizing genes are elevated. This indicates that lipid oxidation may be refractory to cancer cell growth. Studies have shown beneficial effects of carbohydrate restriction in various forms in the treatment of cancer. For example, the use of ketogenic diets, which contain high levels of fat and protein with very low levels of carbohydrates, have shown efficacy in decreasing tumor growth in glioma, colon, prostate, and gastric cancers. A major challenge facing the use of these diets in cancer therapy is that the mechanism by which they show efficacy in cancer remains unclear. By using octanoate, the most well-known ketogenic fatty acid, we are able to drive fatty acid oxidation and ketogenesis to study these processes in proliferating cells. We found that supplementation of octanoate into complete culture medium causes a dramatic, dose-dependent and reversible suppression of proliferation across numerous cell lines and significant changes to anabolic cellular metabolism. Importantly, we have found that ketone production from octanoate had no effect on cell proliferation but that the overall cellular response to the lipid causes inhibition of cell growth.
Nutrients and metabolites are sensed by the cell at many levels and the cellular response to metabolites is critical to proliferation and survival of a cancer cell. One way in which the cell responds to glucose, the major fuel source in cancer cells, is by increasing histone acetylation to promote gene expression. Wellen et al., found that upon glucose addition there was a specific gene expression pattern characterized by the upregulation of genes involved in glucose metabolism. In this way the cell promotes glucose-derived fatty acid synthesis, a rate-limiting process for cancer cell proliferation. This is one way in which the metabolic response to nutrients is integrated into cellular signaling and the epigenome. Remarkably, we have found that lipids can promote a feed-forward mechanism of lipid metabolism by inducing histone acetylation and increasing gene expression of lipid metabolizing genes. We find that upon treatment of cells with octanoate there is an inhibition of both glucose and glutamine metabolism and that octanoate-derived carbon becomes the major fuel source in the cell. We then found that histones were hyperacetylated after octanoate treatment and remarkably, close to 90% of the carbon on histones was octanoate derived. In addition, octanoate is a weak HDAC inhibitor which further promotes octanoate-derived acetyl-CoA being deposited onto histones. A gene array from octanoate treated samples finds that fatty acid metabolism is the top pathway in our gene ontology analysis. This provides evidence that the cell responds to nutrient sources in a specific manner depending on the nature of the carbon source. Finally, we find the most negatively regulated pathway upon octanoate treatment is DNA replication. Consistently, we find that octanoate causes an accumulation of cells in G1 phase of the cell cycle and induction of apoptosis.
Here we describe a mechanism for how fatty acids are sensed and how they communicate with the nucleus to alter gene expression. We show that the cell responds to lipids via a coordinated response to promote lipid metabolism and induce histone acetylation. This feed-forward mechanism of lipid metabolism consists of a reprograming of anabolic metabolism, and promotion of gene expression changes culminating in inhibition of cell growth and apoptosis.
Item Open Access Wnt Protein Signaling Reduces Nuclear Acetyl-CoA Levels to Suppress Gene Expression during Osteoblast Differentiation.(J Biol Chem, 2016-06-17) Karner, Courtney M; Esen, Emel; Chen, Jiakun; Hsu, Fong-Fu; Turk, John; Long, FanxinDevelopmental signals in metazoans play critical roles in inducing cell differentiation from multipotent progenitors. The existing paradigm posits that the signals operate directly through their downstream transcription factors to activate expression of cell type-specific genes, which are the hallmark of cell identity. We have investigated the mechanism through which Wnt signaling induces osteoblast differentiation in an osteoblast-adipocyte bipotent progenitor cell line. Unexpectedly, Wnt3a acutely suppresses the expression of a large number of genes while inducing osteoblast differentiation. The suppressed genes include Pparg and Cebpa, which encode adipocyte-specifying transcription factors and suppression of which is sufficient to induce osteoblast differentiation. The large scale gene suppression induced by Wnt3a corresponds to a global decrease in histone acetylation, an epigenetic modification that is associated with gene activation. Mechanistically, Wnt3a does not alter histone acetyltransferase or deacetylase activities but, rather, decreases the level of acetyl-CoA in the nucleus. The Wnt-induced decrease in histone acetylation is independent of β-catenin signaling but, rather, correlates with suppression of glucose metabolism in the tricarboxylic acid cycle. Functionally, preventing histone deacetylation by increasing nucleocytoplasmic acetyl-CoA levels impairs Wnt3a-induced osteoblast differentiation. Thus, Wnt signaling induces osteoblast differentiation in part through histone deacetylation and epigenetic suppression of an alternative cell fate.