1
|
Yuan VG. Rhythms in Remodeling: Posttranslational Regulation of Bone by the Circadian Clock. Biomedicines 2025; 13:705. [PMID: 40149680 PMCID: PMC11940027 DOI: 10.3390/biomedicines13030705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
The circadian clock is a fundamental timekeeping system that regulates rhythmic biological processes in response to environmental light-dark cycles. In mammals, core clock genes (CLOCK, BMAL1, PER, and CRY) orchestrate these rhythms through transcriptional-translational feedback loops, influencing various physiological functions, including bone remodeling. Bone homeostasis relies on the coordinated activities of osteoblasts, osteoclasts, and osteocytes, with increasing evidence highlighting the role of circadian regulation in maintaining skeletal integrity. Disruptions in circadian rhythms are linked to bone disorders such as osteoporosis. Posttranslational modifications (PTMs), including phosphorylation, acetylation, and ubiquitination, serve as crucial regulators of both circadian mechanisms and bone metabolism. However, the specific role of PTMs in integrating circadian timing with bone remodeling remains underexplored. This review examines the intersection of circadian regulation and PTMs in bone biology, elucidating their impact on bone cell function and homeostasis. Understanding these interactions may uncover novel therapeutic targets for skeletal diseases associated with circadian disruptions.
Collapse
Affiliation(s)
- Vincent G Yuan
- Department of Otolaryngology-Head & Neck Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
2
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. Brain 2024; 147:3471-3486. [PMID: 38554393 PMCID: PMC11449144 DOI: 10.1093/brain/awae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical allodynia. The role of skin mechanoreceptors in the development of mechanical allodynia is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin-keratinocyte specific BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Joy Barrett
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21021, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Catherine Pei-Ju Lu
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - James W Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
Shaikh M, Doshi G. Epigenetic aging in major depressive disorder: Clocks, mechanisms and therapeutic perspectives. Eur J Pharmacol 2024; 978:176757. [PMID: 38897440 DOI: 10.1016/j.ejphar.2024.176757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
Depression, a chronic mental disorder characterized by persistent sadness, loss of interest, and difficulty in daily tasks, impacts millions globally with varying treatment options. Antidepressants, despite their long half-life and minimal effectiveness, leave half of patients undertreated, highlighting the need for new therapies to enhance well-being. Epigenetics, which studies genetic changes in gene expression or cellular phenotype without altering the underlying Deoxyribonucleic Acid (DNA) sequence, is explored in this article. This article delves into the intricate relationship between epigenetic mechanisms and depression, shedding light on how environmental stressors, early-life adversity, and genetic predispositions shape gene expression patterns associated with depression. We have also discussed Histone Deacetylase (HDAC) inhibitors, which enhance cognitive function and mood regulation in depression. Non-coding RNAs, (ncRNAs) such as Long Non-Coding RNAs (lncRNAs) and micro RNA (miRNAs), are highlighted as potential biomarkers for detecting and monitoring major depressive disorder (MDD). This article also emphasizes the reversible nature of epigenetic modifications and their influence on neuronal growth processes, underscoring the dynamic interplay between genetics, environment, and epigenetics in depression development. It explores the therapeutic potential of targeting epigenetic pathways in treating clinical depression. Additionally, it examines clinical findings related to epigenetic clocks and their role in studying depression and biological aging.
Collapse
Affiliation(s)
- Muqtada Shaikh
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India
| | - Gaurav Doshi
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India.
| |
Collapse
|
4
|
Velleuer E, Carlberg C. A Nutrigenomic View on the Premature-Aging Disease Fanconi Anemia. Nutrients 2024; 16:2271. [PMID: 39064714 PMCID: PMC11280142 DOI: 10.3390/nu16142271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Fanconi anemia, a rare disorder with an incidence of 1 in 300,000, is caused by mutations in FANC genes, which affect the repair of DNA interstrand crosslinks. The disease is characterized by congenital malformations, bone marrow failure within the first decade of life, and recurrent squamous cell carcinomas of the oral cavity, esophagus, and anogenital regions starting around age 20. In this review, we propose that Fanconi anemia should be considered a premature-aging syndrome. Interestingly, the onset and severity of the life-limiting clinical features of Fanconi anemia can be influenced by lifestyle choices, such as a healthy diet and physical activity. These factors shape the epigenetic status of at-risk cell types and enhance the competence of the immune system through nutritional signaling. Fanconi anemia may serve as a model for understanding the aging process in the general population, addressing research gaps in its clinical presentation and suggesting prevention strategies. Additionally, we will discuss how the balance of genetic and environmental risk factors-affecting both cancer onset and the speed of aging-is interlinked with signal transduction by dietary molecules. The underlying nutrigenomic principles will offer guidance for healthy aging in individuals with Fanconi anemia as well as for the general population.
Collapse
Affiliation(s)
- Eunike Velleuer
- Department for Cytopathology, Heinrich-Heine-University Düsseldorf, D-40225 Düsseldorf, Germany;
- Department for Pediatric Hemato-Oncology, Helios Children’s Hospital, D-47805 Krefeld, Germany
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, PL-10-748 Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
5
|
Shokeen K, Kumar S. Newcastle disease virus regulates its replication by instigating oxidative stress-driven Sirtuin 7 production. J Gen Virol 2024; 105. [PMID: 38376490 DOI: 10.1099/jgv.0.001961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024] Open
Abstract
Reactive oxygen species (ROS) accumulation inside the cells instigates oxidative stress, activating stress-responsive genes. The viral strategies for promoting stressful conditions and utilizing the induced host proteins to enhance their replication remain elusive. The present work investigates the impact of oxidative stress responses on Newcastle disease virus (NDV) pathogenesis. Here, we show that the progression of NDV infection varies with intracellular ROS levels. Additionally, the results demonstrate that NDV infection modulates the expression of oxidative stress-responsive genes, majorly sirtuin 7 (SIRT7), a NAD+-dependent deacetylase. The modulation of SIRT7 protein, both through overexpression and knockdown, significantly impacts the replication dynamics of NDV in DF-1 cells. The activation of SIRT7 is found to be associated with the positive regulation of cellular protein deacetylation. Lastly, the results suggested that NDV-driven SIRT7 alters NAD+ metabolism in vitro and in ovo. We concluded that the elevated expression of NDV-mediated SIRT7 protein with enhanced activity metabolizes the NAD+ to deacetylase the host proteins, thus contributing to high virus replication.
Collapse
Affiliation(s)
- Kamal Shokeen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
6
|
Yuan M, Yang B, Rothschild G, Mann JJ, Sanford LD, Tang X, Huang C, Wang C, Zhang W. Epigenetic regulation in major depression and other stress-related disorders: molecular mechanisms, clinical relevance and therapeutic potential. Signal Transduct Target Ther 2023; 8:309. [PMID: 37644009 PMCID: PMC10465587 DOI: 10.1038/s41392-023-01519-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/14/2023] [Accepted: 05/31/2023] [Indexed: 08/31/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic, generally episodic and debilitating disease that affects an estimated 300 million people worldwide, but its pathogenesis is poorly understood. The heritability estimate of MDD is 30-40%, suggesting that genetics alone do not account for most of the risk of major depression. Another factor known to associate with MDD involves environmental stressors such as childhood adversity and recent life stress. Recent studies have emerged to show that the biological impact of environmental factors in MDD and other stress-related disorders is mediated by a variety of epigenetic modifications. These epigenetic modification alterations contribute to abnormal neuroendocrine responses, neuroplasticity impairment, neurotransmission and neuroglia dysfunction, which are involved in the pathophysiology of MDD. Furthermore, epigenetic marks have been associated with the diagnosis and treatment of MDD. The evaluation of epigenetic modifications holds promise for further understanding of the heterogeneous etiology and complex phenotypes of MDD, and may identify new therapeutic targets. Here, we review preclinical and clinical epigenetic findings, including DNA methylation, histone modification, noncoding RNA, RNA modification, and chromatin remodeling factor in MDD. In addition, we elaborate on the contribution of these epigenetic mechanisms to the pathological trait variability in depression and discuss how such mechanisms can be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Biao Yang
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Radiology, Columbia University, New York, NY, 10032, USA
| | - Larry D Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuang Wang
- Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Medical Big Data Center, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Mishra S, Welch N, Karthikeyan M, Bellar A, Musich R, Singh SS, Zhang D, Sekar J, Attaway A, Chelluboyina AK, Lorkowski SW, Roychowdhury S, Li L, Willard B, Smith JD, Hoppel C, Vachharajani V, Kumar A, Dasarathy S. Dysregulated cellular redox status during hyperammonemia causes mitochondrial dysfunction and senescence by inhibiting sirtuin-mediated deacetylation. Aging Cell 2023; 22:e13852. [PMID: 37101412 PMCID: PMC10352558 DOI: 10.1111/acel.13852] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/28/2023] Open
Abstract
Perturbed metabolism of ammonia, an endogenous cytotoxin, causes mitochondrial dysfunction, reduced NAD+ /NADH (redox) ratio, and postmitotic senescence. Sirtuins are NAD+ -dependent deacetylases that delay senescence. In multiomics analyses, NAD metabolism and sirtuin pathways are enriched during hyperammonemia. Consistently, NAD+ -dependent Sirtuin3 (Sirt3) expression and deacetylase activity were decreased, and protein acetylation was increased in human and murine skeletal muscle/myotubes. Global acetylomics and subcellular fractions from myotubes showed hyperammonemia-induced hyperacetylation of cellular signaling and mitochondrial proteins. We dissected the mechanisms and consequences of hyperammonemia-induced NAD metabolism by complementary genetic and chemical approaches. Hyperammonemia inhibited electron transport chain components, specifically complex I that oxidizes NADH to NAD+ , that resulted in lower redox ratio. Ammonia also caused mitochondrial oxidative dysfunction, lower mitochondrial NAD+ -sensor Sirt3, protein hyperacetylation, and postmitotic senescence. Mitochondrial-targeted Lactobacillus brevis NADH oxidase (MitoLbNOX), but not NAD+ precursor nicotinamide riboside, reversed ammonia-induced oxidative dysfunction, electron transport chain supercomplex disassembly, lower ATP and NAD+ content, protein hyperacetylation, Sirt3 dysfunction and postmitotic senescence in myotubes. Even though Sirt3 overexpression reversed ammonia-induced hyperacetylation, lower redox status or mitochondrial oxidative dysfunction were not reversed. These data show that acetylation is a consequence of, but is not the mechanism of, lower redox status or oxidative dysfunction during hyperammonemia. Targeting NADH oxidation is a potential approach to reverse and potentially prevent ammonia-induced postmitotic senescence in skeletal muscle. Since dysregulated ammonia metabolism occurs with aging, and NAD+ biosynthesis is reduced in sarcopenia, our studies provide a biochemical basis for cellular senescence and have relevance in multiple tissues.
Collapse
Affiliation(s)
- Saurabh Mishra
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Nicole Welch
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
- Departments of Gastroenterology and HepatologyCleveland ClinicClevelandOhioUSA
| | - Manikandan Karthikeyan
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Annette Bellar
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Ryan Musich
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Shashi Shekhar Singh
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Dongmei Zhang
- Proteomics and Metabolomics coreLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Jinendiran Sekar
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Amy H. Attaway
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
- Departments of Pulmonary MedicineCleveland ClinicClevelandOhioUSA
| | - Aruna Kumar Chelluboyina
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Shuhui Wang Lorkowski
- Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Sanjoy Roychowdhury
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Ling Li
- Proteomics and Metabolomics coreLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Belinda Willard
- Proteomics and Metabolomics coreLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Jonathan D. Smith
- Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Charles L. Hoppel
- Department of PharmacologyCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Vidula Vachharajani
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
- Critical Care Medicine, Respiratory Institute, Cleveland ClinicClevelandOhioUSA
| | - Avinash Kumar
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Srinivasan Dasarathy
- Department of Inflammation and ImmunityLerner Research Institute, Cleveland ClinicClevelandOhioUSA
- Departments of Gastroenterology and HepatologyCleveland ClinicClevelandOhioUSA
| |
Collapse
|
8
|
Elekofehinti OO, Aladenika YV, Iwaloye O, Okon EIA, Adanlawo IG. Bambusa vulgaris leaves reverse mitochondria dysfunction in diabetic rats through modulation of mitochondria biogenic genes. Horm Mol Biol Clin Investig 2023:hmbci-2022-0053. [PMID: 36591918 DOI: 10.1515/hmbci-2022-0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 12/11/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES There is evidence that mitochondrial dysfunction mediated by hyperglycemia increases the incidence of diabetes and age-related insulin resistance. Thus, maintaining mitochondrial integrity may provide alternative therapeutic approach in diabetes treatment. This study aimed to evaluate the effect of Bambusa vulgaris leaf extract on mitochondrial biogenesis in the pancreas of diabetic rats. METHODS 11 weeks old male rats (n=30) were purchased, and sorted into the following groups: control, diabetic control, diabetes + metformin (100 mg/kg), diabetes + Aq. B. vulgaris (100 mg/kg), diabetes + Aq. B. vulgaris (200 mg/kg), and diabetes + Aq. B. vulgaris (300 mg/kg). Diabetes was induced in the rats by a single dose of 65 mg/kg streptozotocin (STZ). The mRNA expression of genes related to mitochondria biogenesis (pgc-1α, Nrf2, GSK3β, AMPK and SIRT2) and genes of Nrf2-Keap1-ARE signaling pathway were determined by reverse transcriptase polymerase chain reaction. Molecular docking studies including lock and key docking and prime MM-GBSA were incorporated to identify the lead chemical compounds in Bambusa vulgari. RESULTS The results showed that B. vulgaris leaf extract promotes mitochondrial biogenesis via altering the mRNA expression of mitochondrial master regulator pgc-1α, other upstream genes, and the Nrf2-Keap1-ARE antioxidant pathway. Through molecular docking results, cryptochlorogenic acid, hesperidin, orientin, vitexin, scopolin, and neochlorogenic were found as the crucial chemicals in B. vulgaris with the most modulating effect on PGC-1α, AMPK, and GSK3. CONCLUSIONS This study thus suggests that B. vulgaris leaf extract restores the integrity of mitochondria in diabetic rats.
Collapse
Affiliation(s)
- Olusola Olalekan Elekofehinti
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, Federal University of Technology Akure, Akure, Ondo State, Nigeria
| | | | - Opeyemi Iwaloye
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, Federal University of Technology Akure, Akure, Ondo State, Nigeria
| | - Enoabasi Ima-Abasi Okon
- Bioinformatics and Molecular Biology Unit, Department of Biochemistry, Federal University of Technology Akure, Akure, Ondo State, Nigeria
| | - Isaac Gbadura Adanlawo
- Department of Biochemistry, Faculty of Science, Ekiti State University, Ado-Ekiti, Nigeria
| |
Collapse
|
9
|
Dai Y, Lin J, Ren J, Zhu B, Wu C, Yu L. NAD + metabolism in peripheral neuropathic pain. Neurochem Int 2022; 161:105435. [PMID: 36273706 DOI: 10.1016/j.neuint.2022.105435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 10/04/2022] [Accepted: 10/16/2022] [Indexed: 11/07/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an omnipresent metabolite that participates in redox reactions. Multiple NAD+-consuming enzymes are implicated in numerous biological processes, including transcription, signaling, and cell survival. Multiple pieces of evidence have demonstrated that NAD+-consuming enzymes, including poly(ADP-ribose) polymerases (PARPs), sirtuins (SIRTs), and sterile alpha and TIR motif-containing 1 (SARM1), play major roles in peripheral neuropathic pain of various etiologies. These NAD+ consumers primarily participate in peripheral neuropathic pain via mechanisms such as mitochondrial dysfunction, oxidative stress, and inflammation. Furthermore, NAD+ synthase and nicotinamide phosphoribosyltransferase (NAMPT) have recently been found to contribute to the regulation of pain. Here, we review the evidence indicating the involvement of NAD+ metabolism in the pathological mechanisms of peripheral neuropathic pain. Advanced understanding of the molecular and cellular mechanisms associated with NAD+ in peripheral neuropathic pain will facilitate the development of novel treatment options for diverse types of peripheral neuropathic pain.
Collapse
Affiliation(s)
- Yi Dai
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Jiaqi Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Jinxuan Ren
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Bin Zhu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Chengwei Wu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Lina Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China.
| |
Collapse
|
10
|
Yan L, Guo MS, Zhang Y, Yu L, Wu JM, Tang Y, Ai W, Zhu FD, Law BYK, Chen Q, Yu CL, Wong VKW, Li H, Li M, Zhou XG, Qin DL, Wu AG. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence, Advances, and Opportunities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5288698. [PMID: 35237381 PMCID: PMC8885204 DOI: 10.1155/2022/5288698] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by the progressive degeneration of neurons. Although the etiology and pathogenesis of neurodegenerative diseases have been studied intensively, the mechanism is still in its infancy. In general, most neurodegenerative diseases share common molecular mechanisms, and multiple risks interact and promote the pathologic process of neurogenerative diseases. At present, most of the approved drugs only alleviate the clinical symptoms but fail to cure neurodegenerative diseases. Numerous studies indicate that dietary plant polyphenols are safe and exhibit potent neuroprotective effects in various neurodegenerative diseases. However, low bioavailability is the biggest obstacle for polyphenol that largely limits its adoption from evidence into clinical practice. In this review, we summarized the widely recognized mechanisms associated with neurodegenerative diseases, such as misfolded proteins, mitochondrial dysfunction, oxidative damage, and neuroinflammatory responses. In addition, we summarized the research advances about the neuroprotective effect of the most widely reported dietary plant polyphenols. Moreover, we discussed the current clinical study and application of polyphenols and the factors that result in low bioavailability, such as poor stability and low permeability across the blood-brain barrier (BBB). In the future, the improvement of absorption and stability, modification of structure and formulation, and the combination therapy will provide more opportunities from the laboratory into the clinic for polyphenols. Lastly, we hope that the present review will encourage further researches on natural dietary polyphenols in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Min-Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yue Zhang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Wei Ai
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Qi Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Department of Nursing, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hua Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mao Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
11
|
Ji L, Jiang W, Liu D, Hou K. Effect of SIRT1 on white matter neural network in adolescent patients with depression. Front Psychiatry 2022; 13:966315. [PMID: 36177213 PMCID: PMC9513552 DOI: 10.3389/fpsyt.2022.966315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND This study aimed to explore the correlation between the rs12415800 polymorphism of the silent information regulator 1 (SIRT1) gene and the white matter neural circuit in adolescent patients with depression. METHODS We enrolled 119 participants, comprising 59 adolescent patients with depression and 60 matched healthy controls for analysis. Patients were further subdivided based on genotype; GG, AG, and AA, with G representing the wild type gene, and A representing the A allele at rs12415800. RESULTS We found that: (1) lower anisotropy fraction (FA) values in the left cingulate fasciculus and left anterior thalamus radiation in the AG/AA genotype were more likely to be affected by depression. (2) The FA values of the right inferior occipital-frontal fasciculus, right corticospinal tract, right inferior longitudinal fasciculus, and right superior longitudinal fasciculus regions in the depression (AG/AA) group were lower than in the depression (GG) group. (3) FA values of the right inferior occipital-frontal fasciculus left corticospinal tract, right inferior longitudinal fasciculus, left anterior thalamus radiation, right superior longitudinal fasciculus, left inferior longitudinal fasciculus, left uncinate fasciculus, and right anterior thalamus radiation in the depression (GG) group were lower than the control (GG) group. CONCLUSIONS The polymorphism locus of the SIRT1 gene rs12415800 may be related to changes in the microstructure of white matter fiber tracts, and patients carrying the A allele (AG/AA) have more changes in the white matter than those with the non-A allele (GG).
Collapse
Affiliation(s)
- Ling Ji
- Department of Clinical Psychology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Wen Jiang
- Outpatient Department, The General Hospital of Western Theater Command, Chengdu, China
| | - Daiyan Liu
- People's Liberation Army of China (PLA) Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Kaiwen Hou
- Outpatient Department, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
12
|
Poitras TM, Munchrath E, Zochodne DW. Neurobiological Opportunities in Diabetic Polyneuropathy. Neurotherapeutics 2021; 18:2303-2323. [PMID: 34935118 PMCID: PMC8804062 DOI: 10.1007/s13311-021-01138-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 12/29/2022] Open
Abstract
This review highlights a selection of potential translational directions for the treatment of diabetic polyneuropathy (DPN) currently irreversible and without approved interventions beyond pain management. The list does not include all diabetic targets that have been generated over several decades of research but focuses on newer work. The emphasis is firstly on approaches that support the viability and growth of peripheral neurons and their ability to withstand a barrage of diabetic alterations. We include a section describing Schwann cell targets and finally how mitochondrial damage has been a common element in discussing neuropathic damage. Most of the molecules and pathways described here have not yet reached clinical trials, but many trials have been negative to date. Nonetheless, these failures clear the pathway for new thoughts over reversing DPN.
Collapse
Affiliation(s)
- Trevor M Poitras
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Easton Munchrath
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Douglas W Zochodne
- Peripheral Nerve Research Laboratory, Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of Alberta, 7-132A Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada.
| |
Collapse
|
13
|
González‐Arzola K, Guerra‐Castellano A, Rivero‐Rodríguez F, Casado‐Combreras MÁ, Pérez‐Mejías G, Díaz‐Quintana A, Díaz‐Moreno I, De la Rosa MA. Mitochondrial cytochrome c shot towards histone chaperone condensates in the nucleus. FEBS Open Bio 2021; 11:2418-2440. [PMID: 33938164 PMCID: PMC8409293 DOI: 10.1002/2211-5463.13176] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Despite mitochondria being key for the control of cell homeostasis and fate, their role in DNA damage response is usually just regarded as an apoptotic trigger. However, growing evidence points to mitochondrial factors modulating nuclear functions. Remarkably, after DNA damage, cytochrome c (Cc) interacts in the cell nucleus with a variety of well-known histone chaperones, whose activity is competitively inhibited by the haem protein. As nuclear Cc inhibits the nucleosome assembly/disassembly activity of histone chaperones, it might indeed affect chromatin dynamics and histone deposition on DNA. Several histone chaperones actually interact with Cc Lys residues through their acidic regions, which are also involved in heterotypic interactions leading to liquid-liquid phase transitions responsible for the assembly of nuclear condensates, including heterochromatin. This relies on dynamic histone-DNA interactions that can be modulated by acetylation of specific histone Lys residues. Thus, Cc may have a major regulatory role in DNA repair by fine-tuning nucleosome assembly activity and likely nuclear condensate formation.
Collapse
Affiliation(s)
- Katiuska González‐Arzola
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Alejandra Guerra‐Castellano
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Francisco Rivero‐Rodríguez
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Miguel Á. Casado‐Combreras
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Gonzalo Pérez‐Mejías
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Antonio Díaz‐Quintana
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Irene Díaz‐Moreno
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| | - Miguel A. De la Rosa
- Institute for Chemical Research (IIQ)Scientific Research Centre Isla de la Cartuja (cicCartuja)University of Seville – CSICSpain
| |
Collapse
|
14
|
Pandey M, Bansal S, Bar S, Yadav AK, Sokol NS, Tennessen JM, Kapahi P, Chawla G. miR-125-chinmo pathway regulates dietary restriction-dependent enhancement of lifespan in Drosophila. eLife 2021; 10:62621. [PMID: 34100717 PMCID: PMC8233039 DOI: 10.7554/elife.62621] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 06/07/2021] [Indexed: 01/06/2023] Open
Abstract
Dietary restriction (DR) extends healthy lifespan in diverse species. Age and nutrient-related changes in the abundance of microRNAs (miRNAs) and their processing factors have been linked to organismal longevity. However, the mechanisms by which they modulate lifespan and the tissue-specific role of miRNA-mediated networks in DR-dependent enhancement of lifespan remains largely unexplored. We show that two neuronally enriched and highly conserved microRNAs, miR-125 and let-7 mediate the DR response in Drosophila melanogaster. Functional characterization of miR-125 demonstrates its role in neurons while its target chinmo acts both in neurons and the fat body to modulate fat metabolism and longevity. Proteomic analysis revealed that Chinmo exerts its DR effects by regulating the expression of FATP, CG2017, CG9577, CG17554, CG5009, CG8778, CG9527, and FASN1. Our findings identify miR-125 as a conserved effector of the DR pathway and open the avenue for this small RNA molecule and its downstream effectors to be considered as potential drug candidates for the treatment of late-onset diseases and biomarkers for healthy aging in humans.
Collapse
Affiliation(s)
- Manish Pandey
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, India
| | - Sakshi Bansal
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, India
| | - Sudipta Bar
- Buck Institute for Research on Aging, Novato, United States
| | - Amit Kumar Yadav
- Translational Health Science and Technology Institute, Faridabad, India
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, United States
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, United States
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, United States
| | - Geetanjali Chawla
- RNA Biology Laboratory, Regional Centre for Biotechnology, Faridabad, India
| |
Collapse
|
15
|
Sarikhani M, Garbern JC, Ma S, Sereda R, Conde J, Krähenbühl G, Escalante GO, Ahmed A, Buenrostro JD, Lee RT. Sustained Activation of AMPK Enhances Differentiation of Human iPSC-Derived Cardiomyocytes via Sirtuin Activation. Stem Cell Reports 2020; 15:498-514. [PMID: 32649901 PMCID: PMC7419706 DOI: 10.1016/j.stemcr.2020.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022] Open
Abstract
Recent studies suggest that metabolic regulation may improve differentiation of cardiomyocytes derived from induced pluripotent stem cells (iPSCs). AMP-activated protein kinase (AMPK) is a master regulator of metabolic activities. We investigated whether AMPK participates in iPSC-derived cardiomyocyte differentiation. We observed that AMPK phosphorylation at Thr172 increased at day 9 but then decreased after day 11 of differentiation to cardiomyocytes. Inhibition of AMPK with compound C significantly reduced mRNA and protein expression of cardiac troponins TNNT2 and TNNI3. Moreover, sustained AMPK activation using AICAR from days 9 to 14 of differentiation increased mRNA and protein expression of both TNNT2 and TNNI3. AICAR decreased acetylation of histone 3 at Lys9 and 56 and histone 4 at Lys16 (known target sites for nuclear-localized sirtuins [SIRT1, SIRT6]), suggesting that AMPK activation enhances sirtuin activity. Sustained AMPK activation during days 9–14 of differentiation induces sirtuin-mediated histone deacetylation and may enhance cardiomyocyte differentiation from iPSCs. iPSC-derived cardiomyocytes transiently increased AMPK phosphorylation at Thr172 Chemical inhibition of AMPK with compound C decreased TNNI3 and TNNT2 expression Sustained activation of AMPK using AICAR increased expression of TNNT2 and TNNI3 AICAR decreased acetylation of histones H3 (at Lys9 and Lys56) and H4 (at Lys16).
Collapse
Affiliation(s)
- Mohsen Sarikhani
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Jessica C Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Sai Ma
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Biology and Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rebecca Sereda
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Jeffrey Conde
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Guido Krähenbühl
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Gabriela O Escalante
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Aishah Ahmed
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Jason D Buenrostro
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA; Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Bossone KA, Ellis JA, Holaska JM. Histone acetyltransferase inhibition rescues differentiation of emerin-deficient myogenic progenitors. Muscle Nerve 2020; 62:128-136. [PMID: 32304242 DOI: 10.1002/mus.26892] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 03/05/2020] [Accepted: 04/07/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Emery-Dreifuss muscular dystrophy (EDMD) is a disease characterized by skeletal muscle wasting, major tendon contractures, and cardiac conduction defects. Mutations in the gene encoding emerin cause EDMD1. Our previous studies suggested that emerin activation of histone deacetylase 3 (HDAC3) to reduce histone 4-lysine 5 (H4K5) acetylation (ac) is important for myogenic differentiation. METHODS Pharmacological inhibitors (Nu9056, L002) of histone acetyltransferases targeting acetylated H4K5 were used to test whether increased acetylated H4K5 was responsible for the impaired differentiation seen in emerin-deficient myogenic progenitors. RESULTS Nu9056 and L002 rescued impaired differentiation in emerin deficiency. SRT1720, which inhibits the nicotinamide adenine dinucleotide (NAD)+ -dependent deacetylase sirtuin 1 (SIRT1), failed to rescue myotube formation. DISCUSSION We conclude that emerin regulation of HDAC3 activity to affect H4K5 acetylation dynamics is important for myogenic differentiation. Targeting H4K5ac dynamics represents a potential new strategy for ameliorating the skeletal muscle wasting seen in EDMD1.
Collapse
Affiliation(s)
- Katherine A Bossone
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, New Jersey, United States.,Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia, Pennsylvania, United States
| | - Joseph A Ellis
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia, Pennsylvania, United States
| | - James M Holaska
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, New Jersey, United States.,Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia, Pennsylvania, United States
| |
Collapse
|
17
|
Abdul NS, Nagiah S, Anand K, Chuturgoon AA. Molecular docking and mechanisms of fusaric acid induced mitochondrial sirtuin aberrations in glycolytically and oxidatively poised human hepatocellular carcinoma (HepG2) cells. Toxicon 2019; 173:48-56. [PMID: 31778683 DOI: 10.1016/j.toxicon.2019.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/05/2019] [Accepted: 11/24/2019] [Indexed: 10/25/2022]
Abstract
Fusaric acid (FA) is a ubiquitous yet neglected mycotoxin. The toxicity of FA is associated with mitochondrial dysfunction and oxidative stress. Sirtuins (SIRTs) are key mediators of cell stress responses through deacetylation of antioxidant, mitochondrial maintenance and energy metabolism proteins. Dietary bioactive compounds have profound effects on SIRT activity, however little is known regarding common foodborne toxins and SIRTs. In this study the interaction of FA with mitochondrial SIRTs - SIRT3 and SIRT5, were firstly studied by molecular docking. Thereafter we substantiated the in silico findings by investigating the effect of FA on expression profiles of SIRT3 and SIRT5, and transcriptional and post-transcriptional regulators, PGC-1α and miRNA-30c using western blots and qPCR in vitro. FA was predicted to bind to the active site of SIRT3 and SIRT5 having implications for biological activity. Furthermore, protein expression of SIRT3 and SIRT5 was down-regulated despite elevated mRNA levels. Further experimentation revealed post-transcriptional regulation of both SIRTs as evidenced by elevated miRNA-30c despite induction of PGC-1α. This study highlights the potential of a diet contaminated with FA to dysregulate mitochondrial specific proteins that can lead to initiation and progression of sirtuin related diseases including cancer and insulin resistance.
Collapse
Affiliation(s)
- Naeem Sheik Abdul
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Savania Nagiah
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Krisnan Anand
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
18
|
Di Malta C, Cinque L, Settembre C. Transcriptional Regulation of Autophagy: Mechanisms and Diseases. Front Cell Dev Biol 2019; 7:114. [PMID: 31312633 PMCID: PMC6614182 DOI: 10.3389/fcell.2019.00114] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022] Open
Abstract
Macro (Autophagy) is a catabolic process that relies on the cooperative function of two organelles: the lysosome and the autophagosome. The recent discovery of a transcriptional gene network that co-regulates the biogenesis and function of these two organelles, and the identification of transcription factors, miRNAs and epigenetic regulators of autophagy, demonstrated that this catabolic process is controlled by both transcriptional and post-transcriptional mechanisms. In this review article, we discuss the nuclear events that control autophagy, focusing particularly on the role of the MiT/TFE transcription factor family. In addition, we will discuss evidence suggesting that the transcriptional regulation of autophagy could be targeted for the treatment of human genetic diseases, such as lysosomal storage disorders (LSDs) and neurodegeneration.
Collapse
Affiliation(s)
- Chiara Di Malta
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Medical and Translational Sciences, University of Naples Federico II, Naples, Italy
| | - Laura Cinque
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Medical and Translational Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
19
|
Cakouros D, Gronthos S. Epigenetic Regulation of Bone Marrow Stem Cell Aging: Revealing Epigenetic Signatures associated with Hematopoietic and Mesenchymal Stem Cell Aging. Aging Dis 2019; 10:174-189. [PMID: 30705777 PMCID: PMC6345334 DOI: 10.14336/ad.2017.1213] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022] Open
Abstract
In this review we explore the importance of epigenetics as a contributing factor for aging adult stem cells. We summarize the latest findings of epigenetic factors deregulated as adult stem cells age and the consequence on stem cell self-renewal and differentiation, with a focus on adult stem cells in the bone marrow. With the latest whole genome bisulphite sequencing and chromatin immunoprecipitations we are able to decipher an emerging pattern common for adult stem cells in the bone marrow niche and how this might correlate to epigenetic enzymes deregulated during aging. We begin by briefly discussing the initial observations in yeast, drosophila and Caenorhabditis elegans (C. elegans) that led to the breakthrough research that identified the role of epigenetic changes associated with lifespan and aging. We then focus on adult stem cells, specifically in the bone marrow, which lends strong support for the deregulation of DNA methyltransferases, histone deacetylases, acetylates, methyltransferases and demethylases in aging stem cells, and how their corresponding epigenetic modifications influence gene expression and the aging phenotype. Given the reversible nature of epigenetic modifications we envisage “epi” targeted therapy as a means to reprogram aged stem cells into their younger counterparts.
Collapse
Affiliation(s)
- Dimitrios Cakouros
- 1Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,2South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Stan Gronthos
- 1Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,2South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
20
|
Uchida S, Yamagata H, Seki T, Watanabe Y. Epigenetic mechanisms of major depression: Targeting neuronal plasticity. Psychiatry Clin Neurosci 2018; 72:212-227. [PMID: 29154458 DOI: 10.1111/pcn.12621] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/02/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022]
Abstract
Major depressive disorder is one of the most common mental illnesses as it affects more than 350 million people globally. Major depressive disorder is etiologically complex and disabling. Genetic factors play a role in the etiology of major depression. However, identical twin studies have shown high rates of discordance, indicating non-genetic mechanisms as well. For instance, stressful life events increase the risk of depression. Environmental stressors also induce stable changes in gene expression within the brain that may lead to maladaptive neuronal plasticity in regions implicated in disease pathogenesis. Epigenetic events alter the chromatin structure and thus modulate expression of genes that play a role in neuronal plasticity, behavioral response to stress, depressive behaviors, and response to antidepressants. Here, we review new information regarding current understanding of epigenetic events that may impact depression. In particular, we discuss the roles of histone acetylation, DNA methylation, and non-coding RNA. These novel mechanisms of action may lead to new therapeutic strategies for treating major depression.
Collapse
Affiliation(s)
- Shusaku Uchida
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Hirotaka Yamagata
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Tomoe Seki
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Yoshifumi Watanabe
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Premature activation of aging-associated molecular mechanisms is emerging as an important contributor to many diseases, including scleroderma. Among central regulators of the aging process are a group of histone deacetylases called sirtuins (SIRTs). Recent findings implicate these molecules as pathophysiological players in scleroderma skin and lung fibrosis. The goal of this article is to review recent studies on the involvement of SIRTs in scleroderma from the perspective of aging-related molecular mechanisms. RECENT FINDINGS Despite a degree of controversy in this rapidly developing field, the majority of data suggest that SIRT levels are decreased in tissues from patients with scleroderma compared to healthy controls as well as in animal models of scleroderma. Molecular studies reveal several mechanisms through which declining SIRT levels contribute to fibrosis, with the most attention given to modulation of the TGF-β signaling pathway. Activation of SIRTs in cell culture and in animal models elicits antifibrotic effects. Declining SIRT levels and activity are emerging as pathophysiological contributors to scleroderma. Restoration of SIRTs may be therapeutic in patients with scleroderma.
Collapse
|
22
|
Shin AN, Han L, Dasgupta C, Huang L, Yang S, Zhang L. SIRT1 increases cardiomyocyte binucleation in the heart development. Oncotarget 2018; 9:7996-8010. [PMID: 29487709 PMCID: PMC5814276 DOI: 10.18632/oncotarget.23847] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/05/2017] [Indexed: 12/17/2022] Open
Abstract
SIRT1 regulates cell senescence. We investigated a novel role of SIRT1 in the regulation of cardiomyocyte terminal differentiation in the developing heart. Retinoic acid (RA)-induced binucleation of H9c2 cells was associated with increased SIRT1 expression. Inhibition of SIRT1 activity or expression significantly decreased RA-induced binucleation. SIRT1 expression was minimal in the fetal heart and significantly upregulated in the hearts of postnatal day 7 (P7) rat pups. In contrast, heart-specific miR-133a expression was high in the fetal heart but significantly reduced in P7 pup hearts. The miR-133a promoter contains a canonical HRE element and hypoxia upregulated miR-133a gene expression in the heart. SIRT1 mRNA 3′UTR has miR-133a binding sequences and miR-133a and hypoxia suppressed SIRT1 expression in cardiomyocytes. Of importance, inhibition of SIRT1 significantly reduced binucleated cardiomyocytes in the hearts of P7 pups. Taken together, the present study reveals a novel role of SIRT1 and its regulation by miR-133a in cardiomyocyte terminal differentiation of the developing heart, and suggests a potential therapeutic strategy that may impact cardiac function later in life.
Collapse
Affiliation(s)
- Alexandra N Shin
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA.,Department of Biological Sciences, California Baptist University, Riverside, California, USA
| | - Limin Han
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Chiranjib Dasgupta
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lei Huang
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Shumei Yang
- Department of Chemistry and Biochemistry, California State University, San Bernardino, California, USA
| | - Lubo Zhang
- The Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
23
|
Aravamudan B, Thompson M, Sieck GC, Vassallo R, Pabelick CM, Prakash YS. Functional Effects of Cigarette Smoke-Induced Changes in Airway Smooth Muscle Mitochondrial Morphology. J Cell Physiol 2016; 232:1053-1068. [PMID: 27474898 DOI: 10.1002/jcp.25508] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/29/2016] [Indexed: 12/16/2022]
Abstract
Long-term exposure to cigarette smoke (CS) triggers airway hyperreactivity and remodeling, effects that involve airway smooth muscle (ASM). We previously showed that CS destabilizes the networked morphology of mitochondria in human ASM by regulating the expression of mitochondrial fission and fusion proteins via multiple signaling mechanisms. Emerging data link regulation of mitochondrial morphology to cellular structure and function. We hypothesized that CS-induced changes in ASM mitochondrial morphology detrimentally affect mitochondrial function, leading to CS effects on contractility and remodeling. Here, ASM cells were exposed to 1% cigarette smoke extract (CSE) for 48 h to alter mitochondrial fission/fusion, or by inhibiting the fission protein Drp1 or the fusion protein Mfn2. Mitochondrial function was assessed via changes in bioenergetics or altered rates of proliferation and apoptosis. Our results indicate that both exposure to CS and inhibition of mitochondrial fission/fusion proteins affect mitochondrial function (i.e., energy metabolism, proliferation, and apoptosis) in ASM cells. In vivo, the airways in mice chronically exposed to CS are thickened and fibrotic, and the expression of proteins involved in mitochondrial function is dramatically altered in the ASM of these mice. We conclude that CS-induced changes in mitochondrial morphology (fission/fusion balance) correlate with mitochondrial function, and thus may control ASM proliferation, which plays a central role in airway health. J. Cell. Physiol. 232: 1053-1068, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bharathi Aravamudan
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Michael Thompson
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Gary C Sieck
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Robert Vassallo
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Christina M Pabelick
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Y S Prakash
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, Minnesota
| |
Collapse
|
24
|
Abstract
Diabetic neuropathy is a dying back neurodegenerative disease of the peripheral nervous system where mitochondrial dysfunction has been implicated as an etiological factor. Diabetes (type 1 or type 2) invokes an elevation of intracellular glucose concentration simultaneously with impaired growth factor support by insulin, and this dual alteration triggers a maladaptation in metabolism of adult sensory neurons. The energy sensing pathway comprising the AMP-activated protein kinase (AMPK)/sirtuin (SIRT)/peroxisome proliferator-activated receptor-γ coactivator α (PGC-1α) signaling axis is the target of these damaging changes in nutrient levels, e.g., induction of nutrient stress, and loss of insulin-dependent growth factor support and instigates an aberrant metabolic phenotype characterized by a suppression of mitochondrial oxidative phosphorylation and shift to anaerobic glycolysis. There is discussion of how this loss of mitochondrial function and transition to overreliance on glycolysis contributes to the diminishment of collateral sprouting and axon regeneration in diabetic neuropathy in the context of the highly energy-consuming nerve growth cone.
Collapse
Affiliation(s)
- Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Research Centre, R4046-351 Taché Ave, Winnipeg, Manitoba, R2H 2A6, Canada.
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, R3E 0T6, Canada.
| |
Collapse
|
25
|
Shen YA, Wang CY, Hsieh YT, Chen YJ, Wei YH. Metabolic reprogramming orchestrates cancer stem cell properties in nasopharyngeal carcinoma. Cell Cycle 2015; 14:86-98. [PMID: 25483072 DOI: 10.4161/15384101.2014.974419] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation of tumor cells endowed with self-renewal capacity and are considered as an underlying cause of tumor recurrence and metastasis. The metabolic signatures of CSCs and the mechanisms involved in the regulation of their stem cell-like properties still remain elusive. We utilized nasopharyngeal carcinoma (NPC) CSCs as a model to dissect their metabolic signatures and found that CSCs underwent metabolic shift and mitochondrial resetting distinguished from their differentiated counterparts. In metabolic shift, CSCs showed a greater reliance on glycolysis for energy supply compared with the parental cells. In mitochondrial resetting, the quantity and function of mitochondria of CSCs were modulated by the biogenesis of the organelles, and the round-shaped mitochondria were distributed in a peri-nuclear manner similar to those seen in the stem cells. In addition, we blocked the glycolytic pathway, increased the ROS levels, and depolarized mitochondrial membranes of CSCs, respectively, and examined the effects of these metabolic factors on CSC properties. Intriguingly, the properties of CSCs were curbed when we redirected the quintessential metabolic reprogramming, which indicates that the plasticity of energy metabolism regulated the balance between acquisition and loss of the stemness status. Taken together, we suggest that metabolic reprogramming is critical for CSCs to sustain self-renewal, deter from differentiation and enhance the antioxidant defense mechanism. Characterization of metabolic reprogramming governing CSC properties is paramount to the design of novel therapeutic strategies through metabolic intervention of CSCs.
Collapse
Key Words
- ATP6, ATP synthase 6
- COX, cytochrome c oxidase
- Cu/ZnSOD, copper/zinc superoxide dismutase
- GLUT1, glucose transporter 1
- GPI, glucose-6-phosphate isomerase
- GR, glutathione reductase
- HK, hexokinase
- MnSOD, manganese superoxide dismutase
- ND1, NADH dehydrogenase subunit 1
- PDH, pyruvate dehydrogenase
- PDK, pyruvate dehydrogenase kinase
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1α
- POLG, mitochondrial DNA polymerase gamma
- TFAM, mitochondrial transcription factor A
- cancer stem cells
- metabolic reprogramming
- metabolic shift
- mitochondrial membrane potential
- mitochondrial resetting
- nasopharyngeal carcinoma
- reactive oxygen species
Collapse
Affiliation(s)
- Yao-An Shen
- a Institute of Biochemistry and Molecular Biology ; Taipei , Taiwan
| | | | | | | | | |
Collapse
|
26
|
Lo Iacono L, Visco-Comandini F, Valzania A, Viscomi MT, Coviello M, Giampà A, Roscini L, Bisicchia E, Siracusano A, Troisi A, Puglisi-Allegra S, Carola V. Adversity in childhood and depression: linked through SIRT1. Transl Psychiatry 2015; 5:e629. [PMID: 26327687 PMCID: PMC5068813 DOI: 10.1038/tp.2015.125] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/14/2015] [Accepted: 07/27/2015] [Indexed: 12/21/2022] Open
Abstract
Experiencing an adverse childhood and parental neglect is a risk factor for depression in the adult population. Patients with a history of traumatic childhood develop a subtype of depression that is characterized by earlier onset, poor treatment response and more severe symptoms. The long-lasting molecular mechanisms that are engaged during early traumatic events and determine the risk for depression are poorly understood. In this study, we altered adult depression-like behavior in mice by applying juvenile isolation stress. We found that this behavioral phenotype was associated with a reduction in the levels of the deacetylase sirtuin1 (SIRT1) in the brain and in peripheral blood mononuclear cells. Notably, peripheral blood mRNA expression of SIRT1 predicted the extent of behavioral despair only when depression-like behavior was induced by juvenile--but not adult--stress, implicating SIRT1 in the regulation of adult behavior at early ages. Consistent with this hypothesis, pharmacological modulation of SIRT1 during juvenile age altered the depression-like behavior in naive mice. We also performed a pilot study in humans, in which the blood levels of SIRT1 correlated significantly with the severity of symptoms in major depression patients, especially in those who received less parental care during childhood. On the basis of these novel findings, we propose the involvement of SIRT1 in the long-term consequences of adverse childhood experiences.
Collapse
Affiliation(s)
- L Lo Iacono
- Department of Experimental Neurosciences, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - F Visco-Comandini
- Department of Physiology and Pharmacology, University of Rome ‘La Sapienza,' Rome, Italy
| | - A Valzania
- Department of Experimental Neurosciences, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - M T Viscomi
- Department of Experimental Neurosciences, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - M Coviello
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Giampà
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - L Roscini
- Department of Psychology and ‘Daniel Bovet' Center, University of Rome ‘La Sapienza,' Rome, Italy
| | - E Bisicchia
- Department of Experimental Neurosciences, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - A Siracusano
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Troisi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - S Puglisi-Allegra
- Department of Experimental Neurosciences, IRCSS Fondazione Santa Lucia, Rome, Italy,Department of Psychology and ‘Daniel Bovet' Center, University of Rome ‘La Sapienza,' Rome, Italy
| | - V Carola
- Department of Experimental Neurosciences, IRCSS Fondazione Santa Lucia, Rome, Italy,Department of Experimental Neurosciences, IRCSS Fondazione Santa Lucia, Via Fosso del Fiorano 63, Rome 00143, Italy. E-mail:
| |
Collapse
|
27
|
Qiu G, Li X, Che X, Wei C, He S, Lu J, Jia Z, Pang K, Fan L. SIRT1 is a regulator of autophagy: Implications in gastric cancer progression and treatment. FEBS Lett 2015; 589:2034-42. [PMID: 26049033 DOI: 10.1016/j.febslet.2015.05.042] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/22/2015] [Accepted: 05/27/2015] [Indexed: 12/20/2022]
Abstract
Silent mating type information regulation 1 (SIRT1) is implicated in tumorigenesis through its effect on autophagy. In gastric cancer (GC), SIRT1 is a marker for prognosis and is involved in cell invasion, proliferation, epithelial-mesenchymal transition (EMT) and drug resistance. Autophagy can function as a cell-survival mechanism or lead to cell death during the genesis and treatment of GC. This functionality is determined by factors including the stage of the tumor, cellular context and stress levels. Interestingly, SIRT1 can regulate autophagy through the deacetylation of autophagy-related genes (ATGs) and mediators of autophagy. Taken together, these findings support the need for continued research efforts to understand the mechanisms mediating the development of gastric cancer and unveil new strategies to eradicate this disease.
Collapse
Affiliation(s)
- Guanglin Qiu
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Xiangming Che
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Chao Wei
- Xi'an Health School, Xi'an 710054, Shaanxi Province, China
| | - Shicai He
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Jing Lu
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Zongliang Jia
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Ke Pang
- Shaanxi Friendship Hospital, Xi'an 710068, Shaanxi Province, China
| | - Lin Fan
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China.
| |
Collapse
|
28
|
Ranhotra HS. The orphan estrogen-related receptor alpha and metabolic regulation: new frontiers. J Recept Signal Transduct Res 2015; 35:565-8. [PMID: 26037200 DOI: 10.3109/10799893.2015.1024853] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Metabolic homeostasis during long-term adaptation in animals is primarily achieved by controlling the expression of metabolic genes by a plethora of cellular transcription factors. The nuclear receptor (NR) superfamily in eukaryotes is an assembly of diverse receptors working as transcriptional regulators of multiple genes. The orphan estrogen-related receptor alpha (ERRα) is one such receptor of the NR superfamily with significant influence on numerous metabolic and other genes. Although it is presently unknown as to which endogenous hormones or ligands activate ERRα, nevertheless it regulates a host of genes whose products participate in various metabolic pathways. Studies over the years show new and interesting data that add to the growing knowledge on ERRα and metabolic regulation. For instance, novel findings indicate existence of mTOR/ERRα regulatory axis and also that ERRα control PGC-1α expression which potentially have significant impact on cellular metabolism. Data show that ERRα exerts its metabolic control by regulating the expression of SIRT5 that influences oxygen consumption and ATP generation. Moreover, ERRα has a role in creatine and lactate uptake in skeletal muscle which is important towards energy generation and contraction. This review is focused on the new insights gained into ERRα regulation of metabolism, networks and pathways that have important consequences in maintaining metabolic homeostasis including development of cancer.
Collapse
Affiliation(s)
- Harmit S Ranhotra
- a Orphan Nuclear Receptors Laboratory, Department of Biochemistry , St. Edmund's College , Shillong, Meghalaya , India
| |
Collapse
|
29
|
Chun P. Role of sirtuins in chronic obstructive pulmonary disease. Arch Pharm Res 2014; 38:1-10. [PMID: 25304127 DOI: 10.1007/s12272-014-0494-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 10/05/2014] [Indexed: 01/14/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by airflow limitation that is associated with chronic inflammatory response to noxious particles or gases. The airflow limitation may be explained by hypersecretion of mucus, thickening and fibrosis of small airways and alveolar wall destruction in emphysema. Sirtuins, a group of class III deacetylases, have gained considerable attention for their positive effects on aging-related disease, such as cancer, cardiovascular disease, neurodegenerative diseases, osteoporosis and COPD. Among the seven mammalian sirtuins, SIRT1-SIRT7, SIRT1 and SIRT6 are considered to have protective effects against COPD. In the lungs, SIRT1 inhibits autophagy, cellular senescence, fibrosis, and inflammation by deacetylation of target proteins using NAD(+) as co-substrate and is therefore linked to the redox state. In addition to SIRT1, SIRT6 have also been shown to improve or slow down COPD. SIRT6 is associated with redox state and inhibits cellular senescence and fibrosis. Therefore, activation of SIRT1 and SIRT6 might be an attractive approach for novel therapeutic targets for COPD. The present review describes the protective effects of SIRT1 and SIRT6 against COPD and their target proteins involved in the pathophysiology of COPD.
Collapse
Affiliation(s)
- Pusoon Chun
- College of Pharmacy, Inje University, 197 Inje-ro, Gimhae, Gyeongnam, 621-749, Korea,
| |
Collapse
|
30
|
Ranhotra HS. Estrogen-related receptor alpha and mitochondria: tale of the titans. J Recept Signal Transduct Res 2014; 35:386-90. [DOI: 10.3109/10799893.2014.959592] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Perovic A, Unic A, Dumic J. Recreational scuba diving: negative or positive effects of oxidative and cardiovascular stress? Biochem Med (Zagreb) 2014; 24:235-47. [PMID: 24969917 PMCID: PMC4083575 DOI: 10.11613/bm.2014.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 02/16/2014] [Indexed: 12/22/2022] Open
Abstract
Environmental conditions and increased physical activity during scuba diving are followed by increased production of free radicals and disturbed redox balance. Redox balance disorder is associated with damage of cellular components, changes of cellular signaling pathways and alterations of gene expression. Oxidative stress leads to increased expression of sirtuins (SIRTs), molecules which play an important role in the antioxidant defense, due to their sensitivity to the changes in the redox status and their ability to regulate redox homeostasis. These facts make SIRTs interesting to be considered as molecules affected by scuba diving and in that sense, as potential biomarkers of oxidative status or possible drug targets in reduction of reactive oxygen species (ROS) accumulation. In addition, SIRTs effects through currently known targets make them intriguing molecules which can act positively on health in general and whose expression can be induced by scuba diving.A demanding physical activity, as well as other circumstances present in scuba diving, has the greatest load on the cardiovascular function (CV). The mechanisms of CV response during scuba diving are still unclear, but diving-induced oxidative stress and the increase in SIRTs expression could be an important factor in CV adaptation. This review summarizes current knowledge on scuba diving-induced oxidative and CV stress and describes the important roles of SIRTs in the (patho)physiological processes caused by the redox balance disorder.
Collapse
Affiliation(s)
- Antonija Perovic
- Department of Biochemical and Hematological Laboratory Diagnostics, Dubrovnik General Hospital, Dubrovnik, Croatia
| | | | | |
Collapse
|
32
|
DNA methyltransferase 1(DNMT1) induced the expression of suppressors of cytokine signaling3 (Socs3) in a mouse model of asthma. Mol Biol Rep 2014; 41:4413-24. [PMID: 24599782 DOI: 10.1007/s11033-014-3312-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 02/24/2014] [Indexed: 01/21/2023]
Abstract
DNMT1 is the most important methyltransferase enzyme, involved in the regulation of gene expression and appropriate histone modification. It interact with proliferating cell nuclear antigen (PCNA), SNF2 family member ATP-dependent chromatin remodeling enzyme, cyclin dependent kinases inhibitor, E2F1 transcription factor and HDACs to form a repressor complex known as HDAC complexes. The interaction of DNMT1 with numerous protein suppressors of promoters suggests that the enzyme is a crucial element of the transcription suppression complex. Since the mechanism behind over expression of Socs3 in Asthma is unclear, we study the Epigenetic mode of overexpression of Socs3 in terms of methylation/acetylation/inactivation of HDACs/activation of HATs enzymes in a mouse model of asthma. The results show that low expression of DNMT1 might indirectly induce the expression of Socs3 and HAT, and inhibit the expression of HDACs family. Furthermore knockdown of DNMT1 by siRNA induced expression of Socs3 while knock down of Socs3 by siRNA has no effect on DNMT1 expression. Our result suggests that the over expression of Socs3 is due to the inhibition of HDACs complex and hyperacetylation of histones molecule along with down regulation of DNMT1 gene. In depth study on DNMT1 might be useful for the development of therapeutic drug against asthma/allergic diseases.
Collapse
|
33
|
Genetic variants in NAMPT predict bladder cancer risk and prognosis in individuals from southwest Chinese Han group. Tumour Biol 2013; 35:4031-40. [PMID: 24363084 DOI: 10.1007/s13277-013-1527-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/06/2013] [Indexed: 12/27/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (Nampt) was served as a useful biomarker for tumorigenesis and for the prediction of cancer survival. In the present study, we analyzed the SNPs of the NAMPT gene and their impact on the susceptibility and prognosis for patients with bladder cancer (BC). The rs61330082, rs2505568 and rs9034 were selected and genotyped by polymerase chain reaction (PCR)-restriction fragment length polymorphism (RFLP) method in 407 patients with bladder cancer and 316 ethnicity-matched healthy control subjects. The genotyping method was confirmed by the DNA sequencing analysis. Statistically significant increased bladder cancer risk was found to be associated with the C allele and CC genotype of rs61330082; nevertheless, decreased bladder cancer risk was revealed to be associated with A allele and AT genotype of rs2505568. Stratified analyses revealed the rs61330082 to be statistically associated with increased bladder cancer risk in smokers and increased invasiveness of bladder cancer. The AT heterozygote of rs2505568 may prevent the recurrence of bladder cancer. Kaplan-Meier curves revealed a statistically significant association of rs2505568 with recurrence-free survival for total bladder cancer patients and non-muscle-invasive bladder cancer patients, and a statistically significant association of rs9034 with recurrence-free survival for muscle-invasive bladder cancer patients. Multiple Cox regression analysis identified the rs2505568 as a possible independent prognostic factor for recurrence-free survival in total bladder cancer patients. Our results suggested an important role for NAMPT in the pathogenesis of bladder cancer and SNPs of NAMPT gene might be a novel genetic biomarker for the prognosis of bladder cancer.
Collapse
|
34
|
Füllgrabe J, Klionsky DJ, Joseph B. The return of the nucleus: transcriptional and epigenetic control of autophagy. Nat Rev Mol Cell Biol 2013; 15:65-74. [PMID: 24326622 DOI: 10.1038/nrm3716] [Citation(s) in RCA: 370] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autophagy is a conserved process by which cytoplasmic components are degraded by the lysosome. It is commonly seen as a cytoplasmic event and, until now, nuclear events were not considered of primary importance for this process. However, recent studies have unveiled a transcriptional and epigenetic network that regulates autophagy. The identification of tightly controlled transcription factors (such as TFEB and ZKSCAN3), microRNAs and histone marks (especially acetylated Lys16 of histone 4 (H4K16ac) and dimethylated H3K9 (H3K9me2)) associated with the autophagic process offers an attractive conceptual framework to understand the short-term transcriptional response and potential long-term responses to autophagy.
Collapse
Affiliation(s)
- Jens Füllgrabe
- Department of Oncology Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm 17176, Sweden
| | - Daniel J Klionsky
- Life Sciences Institute and Departments of Molecular, Cellular and Developmental Biology and Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Bertrand Joseph
- Department of Oncology Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm 17176, Sweden
| |
Collapse
|
35
|
Desquiret-Dumas V, Gueguen N, Leman G, Baron S, Nivet-Antoine V, Chupin S, Chevrollier A, Vessières E, Ayer A, Ferré M, Bonneau D, Henrion D, Reynier P, Procaccio V. Resveratrol induces a mitochondrial complex I-dependent increase in NADH oxidation responsible for sirtuin activation in liver cells. J Biol Chem 2013; 288:36662-75. [PMID: 24178296 DOI: 10.1074/jbc.m113.466490] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Resveratrol (RSV) has been shown to be involved in the regulation of energetic metabolism, generating increasing interest in therapeutic use. SIRT1 has been described as the main target of RSV. However, recent reports have challenged the hypothesis of its direct activation by RSV, and the signaling pathways remain elusive. Here, the effects of RSV on mitochondrial metabolism are detailed both in vivo and in vitro using murine and cellular models and isolated enzymes. We demonstrate that low RSV doses (1-5 μM) directly stimulate NADH dehydrogenases and, more specifically, mitochondrial complex I activity (EC50 ∼1 μM). In HepG2 cells, this complex I activation increases the mitochondrial NAD(+)/NADH ratio. This higher NAD(+) level initiates a SIRT3-dependent increase in the mitochondrial substrate supply pathways (i.e. the tricarboxylic acid cycle and fatty acid oxidation). This effect is also seen in liver mitochondria of RSV-fed animals (50 mg/kg/day). We conclude that the increase in NADH oxidation by complex I is a crucial event for SIRT3 activation by RSV. Our results open up new perspectives in the understanding of the RSV signaling pathway and highlight the critical importance of RSV doses used for future clinical trials.
Collapse
|
36
|
Koen YM, Sarma D, Hajovsky H, Galeva NA, Williams TD, Staudinger JL, Hanzlik RP. Protein targets of thioacetamide metabolites in rat hepatocytes. Chem Res Toxicol 2013; 26:564-74. [PMID: 23465048 PMCID: PMC3710294 DOI: 10.1021/tx400001x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Thioacetamide (TA) has long been known as a hepatotoxicant whose bioactivation requires S-oxidation to thioacetamide S-oxide (TASO) and then to the very reactive S,S-dioxide (TASO2). The latter can tautomerize to form acylating species capable of covalently modifying cellular nucleophiles including phosphatidylethanolamine (PE) lipids and protein lysine side chains. Isolated hepatocytes efficiently oxidize TA to TASO but experience little covalent binding or cytotoxicity because TA is a very potent inhibitor of the oxidation of TASO to TASO2. However, hepatocytes treated with TASO show extensive covalent binding to both lipids and proteins accompanied by extensive cytotoxicity. In this work, we treated rat hepatocytes with [(14)C]-TASO and submitted the mitochondrial, microsomal, and cytosolic fractions to 2DGE, which revealed a total of 321 radioactive protein spots. To facilitate the identification of target proteins and adducted peptides, we also treated cells with a mixture of TASO/[(13)C2D3]-TASO. Using a combination of 1DGE- and 2DGE-based proteomic approaches, we identified 187 modified peptides (174 acetylated, 50 acetimidoylated, and 37 in both forms) from a total of 88 nonredundant target proteins. Among the latter, 57 are also known targets of at least one other hepatotoxin. The formation of both amide- and amidine-type adducts to protein lysine side chains is in contrast to the exclusive formation of amidine-type adducts with PE phospholipids. Thiobenzamide (TB) undergoes the same two-step oxidative bioactivation as TA, and it also gives rise to both amide and amidine adducts on protein lysine side chains but only amidine adducts to PE lipids. Despite their similarity in functional group chemical reactivity, only 38 of 62 known TB target proteins are found among the 88 known targets of TASO. The potential roles of protein modification by TASO in triggering cytotoxicity are discussed in terms of enzyme inhibition, protein folding, and chaperone function, and the emerging role of protein acetylation in intracellular signaling and the regulation of biochemical pathways.
Collapse
Affiliation(s)
- Yakov M. Koen
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045
| | - Diganta Sarma
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045
| | - Heather Hajovsky
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045
| | - Nadezhda A. Galeva
- Mass Spectrometry Laboratory, The University of Kansas, Lawrence, Kansas 66045
| | - Todd D. Williams
- Mass Spectrometry Laboratory, The University of Kansas, Lawrence, Kansas 66045
| | - Jeffrey L. Staudinger
- Department of Pharmacology & Toxicology, The University of Kansas, Lawrence, Kansas 66045
| | - Robert P. Hanzlik
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
37
|
Liu J, Wu X, Wang X, Zhang Y, Bu P, Zhang Q, Jiang F. Global Gene Expression Profiling Reveals Functional Importance of Sirt2 in Endothelial Cells under Oxidative Stress. Int J Mol Sci 2013; 14:5633-49. [PMID: 23478437 PMCID: PMC3634502 DOI: 10.3390/ijms14035633] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/22/2013] [Accepted: 02/28/2013] [Indexed: 01/30/2023] Open
Abstract
The NAD+-dependent deacetylases Sirt1 and Sirt2 mediate cellular stress responses and are highly expressed in vascular endothelial cells. In contrast to the well-documented protective actions of Sirt1, the role of endothelial Sirt2 remains unknown. Using cDNA microarray and PCR validation, we examined global gene expression changes in response to Sirt2 knock down in primary human umbilical vein endothelial cells under oxidative stress. We found that Sirt2 knock down changed expression of 340 genes, which are mainly involved in cellular processes including actin binding, cellular amino acid metabolic process, transmembrane receptor protein serine/threonine kinase signaling, ferrous iron transport, protein transport and localization, cell morphogenesis, and functions associated with endosome membrane and the trans-Golgi network. These genes and associated functions were largely non-overlapping with those altered by Sirt1 knock down. Moreover, we showed that pharmacological inhibition of Sirt2 attenuated oxidant-induced cell toxicity in endothelial cells. These suggest that Sirt2 is functionally important in endothelial cells under oxidative stress, and may have a primarily distinct role as compared to Sirt1. Our results may provide a basis for future studies aiming to dissect the specific signaling pathway(s) that mediates specific Sirt2 functions in endothelial cells.
Collapse
Affiliation(s)
- Junni Liu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, Shandong, China; E-Mails: (J.L.); (X.Wu.); (X.Wa.); (Y.Z.)
- Department of Cardiology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
| | - Xiao Wu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, Shandong, China; E-Mails: (J.L.); (X.Wu.); (X.Wa.); (Y.Z.)
| | - Xi Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, Shandong, China; E-Mails: (J.L.); (X.Wu.); (X.Wa.); (Y.Z.)
- Department of Cardiology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, Shandong, China; E-Mails: (J.L.); (X.Wu.); (X.Wa.); (Y.Z.)
| | - Peili Bu
- Department of Cardiology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
- Authors to whom correspondence should be addressed; E-Mails: (P.B.); (Q.Z.); (F.J.); Tel.: +86-531-8216-9267 (F.J.); Fax: +86-531-8616-9356 (F.J.)
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, Shandong, China; E-Mails: (J.L.); (X.Wu.); (X.Wa.); (Y.Z.)
- Authors to whom correspondence should be addressed; E-Mails: (P.B.); (Q.Z.); (F.J.); Tel.: +86-531-8216-9267 (F.J.); Fax: +86-531-8616-9356 (F.J.)
| | - Fan Jiang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, Shandong, China; E-Mails: (J.L.); (X.Wu.); (X.Wa.); (Y.Z.)
- Authors to whom correspondence should be addressed; E-Mails: (P.B.); (Q.Z.); (F.J.); Tel.: +86-531-8216-9267 (F.J.); Fax: +86-531-8616-9356 (F.J.)
| |
Collapse
|
38
|
Sakkiah S, Arooj M, Kumar MR, Eom SH, Lee KW. Identification of inhibitor binding site in human sirtuin 2 using molecular docking and dynamics simulations. PLoS One 2013; 8:e51429. [PMID: 23382805 PMCID: PMC3557295 DOI: 10.1371/journal.pone.0051429] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 11/01/2012] [Indexed: 11/19/2022] Open
Abstract
The ability to identify the site of a protein that can bind with high affinity to small, drug-like compounds has been an important goal in drug design. Sirtuin 2 (SIRT2), histone deacetylase protein family, plays a central role in the regulation of various pathways. Hence, identification of drug for SIRT2 has attracted great interest in the drug discovery community. To elucidate the molecular basis of the small molecules interactions to inhibit the SIRT2 function we employed the molecular docking, molecular dynamics simulations, and the molecular mechanism Poisson-Boltzmann/surface area (MM-PBSA) calculations. Five well know inhibitors such as suramin, mol-6, sirtinol, 67, and nf675 were selected to establish the nature of the binding mode of the inhibitors in the SIRT2 active site. The molecular docking and dynamics simulations results revealed that the hydrogen bonds between Arg97 and Gln167 are crucial to inhibit the function of SIRT2. In addition, the MM-PBSA calculations revealed that binding of inhibitors to SIRT2 is mainly driven by van der Waals/non-polar interactions. Although the five inhibitors are very different in structure, shape, and electrostatic potential, they are able to fit in the same binding pocket. These findings from this study provide insights to elucidate the binding pattern of SIRT2 inhibitors and help in the rational structure-based design of novel SIRT2 inhibitors with improved potency and better resistance profile.
Collapse
Affiliation(s)
- Sugunadevi Sakkiah
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Steitz Center for Structural Biology, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Department of Chemistry and Institute of Basic Science, Chonnam National University, Gwangju, South Korea
| | - Mahreen Arooj
- Division of Applied Life Science, Systems and Synthetic Agrobiotech Center, Plant Molecular Biology and Biotechnology Research Center, Research Institute of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Manian Rajesh Kumar
- Department of Chemistry and Institute of Basic Science, Chonnam National University, Gwangju, South Korea
| | - Soo Hyun Eom
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
- Steitz Center for Structural Biology, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Keun Woo Lee
- Division of Applied Life Science, Systems and Synthetic Agrobiotech Center, Plant Molecular Biology and Biotechnology Research Center, Research Institute of Natural Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
39
|
Malireddy S, Kotha SR, Secor JD, Gurney TO, Abbott JL, Maulik G, Maddipati KR, Parinandi NL. Phytochemical antioxidants modulate mammalian cellular epigenome: implications in health and disease. Antioxid Redox Signal 2012; 17:327-39. [PMID: 22404530 PMCID: PMC3353820 DOI: 10.1089/ars.2012.4600] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED In living systems, the mechanisms of inheritance involving gene expression are operated by (i) the traditional model of genetics where the deoxyribonucleic acid (DNA) transcription and messenger ribonucleic acid stability are influenced by the DNA sequences and any aberrations in the primary DNA sequences and (ii) the epigenetic (above genetics) model in which the gene expression is regulated by mechanisms other than the changes in DNA sequences. The widely studied epigenetic alterations include DNA methylation, covalent modification of chromatin structure, state of histone acetylation, and involvement of microribonucleic acids. SIGNIFICANCE Currently, the role of cellular epigenome in health and disease is rapidly emerging. Several factors are known to modulate the epigenome-regulated gene expression that is crucial in several pathophysiological states and diseases in animals and humans. Phytochemicals have occupied prominent roles in human diet and nutrition as protective antioxidants in prevention/protection against several disorders and diseases in humans. RECENT ADVANCES However, it is beginning to surface that the phytochemical phenolic antioxidants such as polyphenols, flavonoids, and nonflavonoid phenols function as potent modulators of the mammalian epigenome-regulated gene expression through regulation of DNA methylation, histone acetylation, and histone deacetylation in experimental models. CRITICAL ISSUES AND FUTURE DIRECTIONS The antioxidant or pro-oxidant actions and their involvement in the epigenome regulation by the phytochemical phenolic antioxidants should be at least established in the cellular models under normal and pathophysiological states. The current review discusses the mechanisms of modulation of the mammalian cellular epigenome by the phytochemical phenolic antioxidants with implications in human diseases.
Collapse
Affiliation(s)
- Smitha Malireddy
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Lipid Signaling, Lipidomics, and Vasculotoxicity Laboratory, Dorothy M. Davis Heart and Lung Research Institute, Colleges of Medicine and Pharmacy, The Ohio State University, Columbus, Ohio
| | - Sainath R. Kotha
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Lipid Signaling, Lipidomics, and Vasculotoxicity Laboratory, Dorothy M. Davis Heart and Lung Research Institute, Colleges of Medicine and Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jordan D. Secor
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Lipid Signaling, Lipidomics, and Vasculotoxicity Laboratory, Dorothy M. Davis Heart and Lung Research Institute, Colleges of Medicine and Pharmacy, The Ohio State University, Columbus, Ohio
| | - Travis O. Gurney
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Lipid Signaling, Lipidomics, and Vasculotoxicity Laboratory, Dorothy M. Davis Heart and Lung Research Institute, Colleges of Medicine and Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jamie L. Abbott
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Lipid Signaling, Lipidomics, and Vasculotoxicity Laboratory, Dorothy M. Davis Heart and Lung Research Institute, Colleges of Medicine and Pharmacy, The Ohio State University, Columbus, Ohio
| | - Gautam Maulik
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Krishna R. Maddipati
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan
| | - Narasimham L. Parinandi
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Lipid Signaling, Lipidomics, and Vasculotoxicity Laboratory, Dorothy M. Davis Heart and Lung Research Institute, Colleges of Medicine and Pharmacy, The Ohio State University, Columbus, Ohio
| |
Collapse
|
40
|
Reichert N, Choukrallah MA, Matthias P. Multiple roles of class I HDACs in proliferation, differentiation, and development. Cell Mol Life Sci 2012; 69:2173-87. [PMID: 22286122 PMCID: PMC11115120 DOI: 10.1007/s00018-012-0921-9] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 01/07/2012] [Accepted: 01/09/2012] [Indexed: 12/19/2022]
Abstract
Class I Histone deacetylases (HDACs) play a central role in controlling cell cycle regulation, cell differentiation, and tissue development. These enzymes exert their function by deacetylating histones and a growing number of non-histone proteins, thereby regulating gene expression and several other cellular processes. Class I HDACs comprise four members: HDAC1, 2, 3, and 8. Deletion and/or overexpression of these enzymes in mammalian systems has provided important insights about their functions and mechanisms of action which are reviewed here. In particular, unique as well as redundant functions have been identified in several paradigms. Studies with small molecule inhibitors of HDACs have demonstrated the medical relevance of these enzymes and their potential as therapeutic targets in cancer and other pathological conditions. Going forward, better understanding the specific role of individual HDACs in normal physiology as well as in pathological settings will be crucial to exploit this protein family as a useful therapeutic target in a range of diseases. Further dissection of the pathways they impinge on and of their targets, in chromatin or otherwise, will form important avenues of research for the future.
Collapse
Affiliation(s)
- Nina Reichert
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, PO Box 2543, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Mohamed-Amin Choukrallah
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, PO Box 2543, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, PO Box 2543, Maulbeerstrasse 66, 4058 Basel, Switzerland
| |
Collapse
|
41
|
Bowlby SC, Thomas MJ, D’Agostino RB, Kridel SJ. Nicotinamide phosphoribosyl transferase (Nampt) is required for de novo lipogenesis in tumor cells. PLoS One 2012; 7:e40195. [PMID: 22768255 PMCID: PMC3387004 DOI: 10.1371/journal.pone.0040195] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 06/02/2012] [Indexed: 11/18/2022] Open
Abstract
Tumor cells have increased metabolic requirements to maintain rapid growth. In particular, a highly lipogenic phenotype is a hallmark of many tumor types, including prostate. Cancer cells also have increased turnover of nicotinamide adenine dinucleotide (NAD+), a coenzyme involved in multiple metabolic pathways. However, a specific role for NAD+ in tumor cell lipogenesis has yet to be described. Our studies demonstrate a novel role for the NAD+-biosynthetic enzyme Nicotinamide phosphoribosyltransferase (Nampt) in maintaining de novo lipogenesis in prostate cancer (PCa) cells. Inhibition of Nampt reduces fatty acid and phospholipid synthesis. In particular, short chain saturated fatty acids and the phosphatidylcholine (PC) lipids into which these fatty acids are incorporated were specifically reduced by Nampt inhibition. Nampt blockade resulted in reduced ATP levels and concomitant activation of AMP-activated protein kinase (AMPK) and phosphorylation of acetyl-CoA carboxylase (ACC). In spite of this, pharmacological inhibition of AMPK was not sufficient to fully restore fatty acid synthesis. Rather, Nampt blockade also induced protein hyperacetylation in PC-3, DU145, and LNCaP cells, which correlated with the observed decreases in lipid synthesis. Moreover, the sirtuin inhibitor Sirtinol, and the simultaneous knockdown of SIRT1 and SIRT3, phenocopied the effects of Nampt inhibition on fatty acid synthesis. Altogether, these data reveal a novel role for Nampt in the regulation of de novo lipogenesis through the modulation of sirtuin activity in PCa cells.
Collapse
Affiliation(s)
- Sarah C. Bowlby
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Michael J. Thomas
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Ralph B. D’Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Steven J. Kridel
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
42
|
Roy Chowdhury SK, Smith DR, Saleh A, Schapansky J, Marquez A, Gomes S, Akude E, Morrow D, Calcutt NA, Fernyhough P. Impaired adenosine monophosphate-activated protein kinase signalling in dorsal root ganglia neurons is linked to mitochondrial dysfunction and peripheral neuropathy in diabetes. ACTA ACUST UNITED AC 2012; 135:1751-66. [PMID: 22561641 DOI: 10.1093/brain/aws097] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Mitochondrial dysfunction occurs in sensory neurons and may contribute to distal axonopathy in animal models of diabetic neuropathy. The adenosine monophosphate-activated protein kinase and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) signalling axis senses the metabolic demands of cells and regulates mitochondrial function. Studies in muscle, liver and cardiac tissues have shown that the activity of adenosine monophosphate-activated protein kinase and PGC-1α is decreased under hyperglycaemia. In this study, we tested the hypothesis that deficits in adenosine monophosphate-activated protein kinase/PGC-1α signalling in sensory neurons underlie impaired axonal plasticity, suboptimal mitochondrial function and development of neuropathy in rodent models of type 1 and type 2 diabetes. Phosphorylation and expression of adenosine monophosphate-activated protein kinase/PGC-1α and mitochondrial respiratory chain complex proteins were downregulated in dorsal root ganglia of both streptozotocin-diabetic rats and db/db mice. Adenoviral-mediated manipulation of endogenous adenosine monophosphate-activated protein kinase activity using mutant proteins modulated neurotrophin-directed neurite outgrowth in cultures of sensory neurons derived from adult rats. Addition of resveratrol to cultures of sensory neurons derived from rats after 3-5 months of streptozotocin-induced diabetes, significantly elevated adenosine monophosphate-activated protein kinase levels, enhanced neurite outgrowth and normalized mitochondrial inner membrane polarization in axons. The bioenergetics profile (maximal oxygen consumption rate, coupling efficiency, respiratory control ratio and spare respiratory capacity) was aberrant in cultured sensory neurons from streptozotocin-diabetic rats and was corrected by resveratrol treatment. Finally, resveratrol treatment for the last 2 months of a 5-month period of diabetes reversed thermal hypoalgesia and attenuated foot skin intraepidermal nerve fibre loss and reduced myelinated fibre mean axonal calibre in streptozotocin-diabetic rats. These data suggest that the development of distal axonopathy in diabetic neuropathy is linked to nutrient excess and mitochondrial dysfunction via defective signalling of the adenosine monophosphate-activated protein kinase/PGC-1α pathway.
Collapse
Affiliation(s)
- Subir K Roy Chowdhury
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Centre, R4023-1 - 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Phillips D, Covian R, Aponte AM, Glancy B, Taylor JF, Chess D, Balaban RS. Regulation of oxidative phosphorylation complex activity: effects of tissue-specific metabolic stress within an allometric series and acute changes in workload. Am J Physiol Regul Integr Comp Physiol 2012; 302:R1034-48. [PMID: 22378775 DOI: 10.1152/ajpregu.00596.2011] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The concentration of mitochondrial oxidative phosphorylation complexes (MOPCs) is tuned to the maximum energy conversion requirements of a given tissue; however, whether the activity of MOPCs is altered in response to acute changes in energy conversion demand is unclear. We hypothesized that MOPCs activity is modulated by tissue metabolic stress to maintain the energy-metabolism homeostasis. Metabolic stress was defined as the observed energy conversion rate/maximum energy conversion rate. The maximum energy conversion rate was assumed to be proportional to the concentration of MOPCs, as determined with optical spectroscopy, gel electrophoresis, and mass spectrometry. The resting metabolic stress of the heart and liver across the range of resting metabolic rates within an allometric series (mouse, rabbit, and pig) was determined from MPOCs content and literature respiratory values. The metabolic stress of the liver was high and nearly constant across the allometric series due to the proportional increase in MOPCs content with resting metabolic rate. In contrast, the MOPCs content of the heart was essentially constant in the allometric series, resulting in an increasing metabolic stress with decreasing animal size. The MOPCs activity was determined in native gels, with an emphasis on Complex V. Extracted MOPCs enzyme activity was proportional to resting metabolic stress across tissues and species. Complex V activity was also shown to be acutely modulated by changes in metabolic stress in the heart, in vivo and in vitro. The modulation of extracted MOPCs activity suggests that persistent posttranslational modifications (PTMs) alter MOPCs activity both chronically and acutely, specifically in the heart. Protein phosphorylation of Complex V was correlated with activity inhibition under several conditions, suggesting that protein phosphorylation may contribute to activity modulation with energy metabolic stress. These data are consistent with the notion that metabolic stress modulates MOPCs activity in the heart.
Collapse
Affiliation(s)
- Darci Phillips
- Laboratory of Cardiac Energetics, NHLBI, NIH, Bethesda, MD 20892-1061, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Webster BR, Lu Z, Sack MN, Scott I. The role of sirtuins in modulating redox stressors. Free Radic Biol Med 2012; 52:281-90. [PMID: 22085655 PMCID: PMC3253188 DOI: 10.1016/j.freeradbiomed.2011.10.484] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/24/2011] [Accepted: 10/25/2011] [Indexed: 12/21/2022]
Abstract
For much of the time since their discovery, the sirtuin family of deacetylase enzymes has been associated with extension of life span. This longevity-promoting capacity in numerous model systems has enabled the sirtuins to gain "celebrity status" in the field of aging research. However, the mechanisms underpinning these changes remain incompletely defined. A general phenotype long associated with aging is the dysregulation of biological systems, which partly occurs via the accumulation of damage over time. One of the major sources of this damage is oxidative stress, which can harm both biological structures and the mechanisms with which they are repaired. It is now becoming clear that the beneficial life-span effects of sirtuins, along with many of their other functions, are closely linked to their ability to regulate systems that control the redox environment. Here we investigate the links between sirtuins and their oxidative/redox environment and review the control mechanisms that are regulated by the activity of sirtuin deacetylase proteins.
Collapse
Affiliation(s)
- Bradley R Webster
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
45
|
The role of SIRT3 in mitochondrial homeostasis and cardiac adaptation to hypertrophy and aging. J Mol Cell Cardiol 2011; 52:520-5. [PMID: 22119802 DOI: 10.1016/j.yjmcc.2011.11.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 11/04/2011] [Accepted: 11/10/2011] [Indexed: 12/16/2022]
Abstract
Although acetyl-modification of protein lysine residues has been recognized for many decades, the appreciation that this post-translational modification is highly prevalent in mitochondria and plays a pivotal regulatory role in mitochondrial function has only become apparent since 2006. The classical biological stressors that modulate mitochondrial protein acetylation include alterations in caloric levels and redox signaling and the major enzyme orchestrating deacetylation is the mitochondrial enriched sirtuin SIRT3. Overall the action of SIRT3 modulates mitochondrial homeostasis and SIRT3 target proteins include mediators of energy metabolism and mitochondrial redox stress adaptive program proteins. Given these effects, it is not surprising that the role of SIRT3 has begun to be implicated in cardiac biology. This review gives a brief overview of sirtuin biology and then focuses on the role of the SIRT3 regulatory program in the control of cardiac hypertrophy and aging. This article is part of a Special Section entitled "Post-translational Modification."
Collapse
|
46
|
Glidden EJ, Gray LG, Vemuru S, Li D, Harris TE, Mayo MW. Multiple site acetylation of Rictor stimulates mammalian target of rapamycin complex 2 (mTORC2)-dependent phosphorylation of Akt protein. J Biol Chem 2011; 287:581-588. [PMID: 22084251 DOI: 10.1074/jbc.m111.304337] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The serine/threonine protein kinase Akt is a critical regulator of cell growth and survival in response to growth factors. A key step in Akt activation is phosphorylation at Ser-473 by the mammalian target of rapamycin (mTOR) complex 2 (mTORC2). Although Rictor is required for the stability and activity of mTORC2, little is known about functional regions or post-translational modifications within Rictor that are responsible for regulating mTORC2. Here, we demonstrate that Rictor contains two distinct central regions critical for mTORC2 function. One we refer to as the stability region because it is critical for interaction with Sin1.1 and LST8, and a second adjacent region is required for multisite acetylation. p300-mediated acetylation of Rictor increases mTORC2 activity toward Akt, whereas site-directed mutants within the acetylation region of Rictor exhibit reduced insulin-like growth factor 1 (IGF-1)-stimulated mTORC2 kinase activity. Inhibition of deacetylases, including the NAD+-dependent sirtuins, promotes Rictor acetylation and IGF-1-mediated Akt phosphorylation. These results suggest that multiple-site acetylation of Rictor signals for increased activation of mTORC2, providing a critical link between nutrient-sensitive deacetylases and mTORC2 signaling to Akt.
Collapse
Affiliation(s)
- Emily J Glidden
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, 22908
| | - Lisa G Gray
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, 22908
| | - Suneil Vemuru
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, 22908
| | - Duo Li
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, 22908
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | - Marty W Mayo
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, 22908.
| |
Collapse
|
47
|
Nadtochiy SM, Yao H, McBurney MW, Gu W, Guarente L, Rahman I, Brookes PS. SIRT1-mediated acute cardioprotection. Am J Physiol Heart Circ Physiol 2011; 301:H1506-12. [PMID: 21856913 DOI: 10.1152/ajpheart.00587.2011] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Overexpression studies have revealed a role for silent information regulator of transcription 1 (SIRT1) lysine deacetylase in cardioprotection against ischemia-reperfusion injury via long-term transcriptional effects. However, short-term SIRT1-mediated lysine deacetylation, within the context of acute cardioprotection, is poorly understood. In this study, the role of SIRT1 in the acute cardioprotective paradigm of first window ischemic preconditioning (IPC) was studied using SIRT1-deficient (SIRT1(+/-)) and SIRT1-overexpressing (SIRT1(+++)) mice. In wild-type hearts, cytosolic lysine deacetylation was observed during IPC, and overacetylation was observed upon pharmacological SIRT1 inhibition. Consistent with a role for SIRT1 in IPC, SIRT1(+/-) hearts could not be preconditioned and exhibited increased cytosolic lysine acetylation. Furthermore, SIRT1(+++) hearts were endogenously protected against ischemia-reperfusion injury and exhibited decreased cytosolic acetylation. Both of these effects in SIRT1(+++) mice were reversed by pharmacological SIRT1 inhibition on an acute timescale. Several downstream targets of SIRT1 were examined, with data suggesting possible roles for endothelial nitric oxide synthase phosphorylation, NF-κB, and stimulation of autophagy. In conclusion, these data suggest that SIRT1, acting on nontranscriptional targets, is required for cardioprotection by acute IPC and that SIRT1-dependent lysine deacetylation occurs during IPC and may play a role in cardioprotective signaling.
Collapse
Affiliation(s)
- Sergiy M Nadtochiy
- Department of Anesthesiology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Abe N, Uchida S, Otsuki K, Hobara T, Yamagata H, Higuchi F, Shibata T, Watanabe Y. Altered sirtuin deacetylase gene expression in patients with a mood disorder. J Psychiatr Res 2011; 45:1106-12. [PMID: 21349544 DOI: 10.1016/j.jpsychires.2011.01.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 01/27/2011] [Accepted: 01/27/2011] [Indexed: 11/17/2022]
Abstract
Sirtuins are a family of NAD+-dependent enzymes that regulate cellular functions through deacetylation of various proteins. Although recent reports have suggested an important role of deacetylases (i.e., histone deacetylases) in mood disorders and antidepressant action, the involvement of sirtuins in the pathophysiology of mood disorders is largely unknown. In this study, we aimed to determine whether there are alterations in sirtuin mRNA expression in peripheral white blood cells of patients with a mood disorder. Also, to examine whether the altered sirtuin mRNA expression is state- or trait-dependent, mood disorder patients who were in a remissive state were assessed. We used quantitative real-time polymerase chain reaction to measure the mRNA levels of seven sirtuin isoforms (SIRT1-7) in peripheral white blood cells of patients with major depressive disorder (MDD) or bipolar disorder (BPD) during depressive and remissive states and in normal healthy subjects. The SIRT1, 2 and 6 mRNA levels in MDD and BPD patients decreased significantly in those who were in a depressive state compared to healthy controls, whereas the expression of those mRNAs in both MDD and BPD of patients in a remissive state were comparable to those in healthy controls. Thus, our data suggest that altered SIRT1, 2 and 6 expression is state-dependent and might be associated with the pathogenesis and/or pathophysiology of mood disorders.
Collapse
Affiliation(s)
- Naoko Abe
- Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Chowdhury SKR, Dobrowsky RT, Fernyhough P. Nutrient excess and altered mitochondrial proteome and function contribute to neurodegeneration in diabetes. Mitochondrion 2011; 11:845-54. [PMID: 21742060 DOI: 10.1016/j.mito.2011.06.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 04/28/2011] [Accepted: 06/24/2011] [Indexed: 01/01/2023]
Abstract
Diabetic neuropathy is a major complication of diabetes that results in the progressive deterioration of the sensory nervous system. Mitochondrial dysfunction has been proposed to play an important role in the pathogenesis of the neurodegeneration observed in diabetic neuropathy. Our recent work has shown that mitochondrial dysfunction occurs in dorsal root ganglia (DRG) sensory neurons in streptozotocin (STZ) induced diabetic rodents. In neurons, the nutrient excess associated with prolonged diabetes may trigger a switching off of AMP kinase (AMPK) and/or silent information regulator T1 (SIRT1) signaling leading to impaired peroxisome proliferator-activated receptor γ coactivator-1 (PGC-1α) expression/activity and diminished mitochondrial activity. This review briefly summarizes the alterations of mitochondrial function and proteome in sensory neurons of STZ-diabetic rodents. We also discuss the possible involvement of AMPK/SIRT/PGC-1α pathway in other diabetic models and different tissues affected by diabetes.
Collapse
Affiliation(s)
- Subir K Roy Chowdhury
- Division of Neurodegenerative Disorders, St Boniface Hospital Research Centre, Winnipeg, MB, Canada R2H 2A6
| | | | | |
Collapse
|
50
|
Shinmura K, Tamaki K, Sano M, Nakashima-Kamimura N, Wolf AM, Amo T, Ohta S, Katsumata Y, Fukuda K, Ishiwata K, Suematsu M, Adachi T. Caloric restriction primes mitochondria for ischemic stress by deacetylating specific mitochondrial proteins of the electron transport chain. Circ Res 2011; 109:396-406. [PMID: 21700931 DOI: 10.1161/circresaha.111.243097] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
RATIONALE Caloric restriction (CR) confers cardioprotection against ischemia/reperfusion injury. However, the exact mechanism(s) underlying CR-induced cardioprotection remain(s) unknown. Recent evidence indicates that Sirtuins, NAD(+)-dependent deacetylases, regulate various favorable aspects of the CR response. Thus, we hypothesized that deacetylation of specific mitochondrial proteins during CR preserves mitochondrial function and attenuates production of reactive oxygen species during ischemia/reperfusion. OBJECTIVE The objectives of the present study were (1) to investigate the effect of CR on mitochondrial function and mitochondrial proteome and (2) to investigate what molecular mechanisms mediate CR-induced cardioprotection. METHODS AND RESULTS Male 26-week-old Fischer344 rats were randomly divided into ad libitum-fed and CR (40% reduction) groups for 6 months. No change was observed in basal mitochondrial function, but CR preserved postischemic mitochondrial respiration and attenuated postischemic mitochondrial H(2)O(2) production. CR decreased the level of acetylated mitochondrial proteins that were associated with enhanced Sirtuin activity in the mitochondrial fraction. We confirmed a significant decrease in the acetylated forms of NDUFS1 and cytochrome bc1 complex Rieske subunit in the CR heart. Low-dose resveratrol treatment mimicked the effect of CR on deacetylating them and attenuated reactive oxygen species production during anoxia/reoxygenation in cultured cardiomyocytes without changing the expression levels of manganese superoxide dismutase. Treatment with nicotinamide completely abrogated the effect of low-dose resveratrol. CONCLUSIONS These results strongly suggest that CR primes mitochondria for stress resistance by deacetylating specific mitochondrial proteins of the electron transport chain. Targeted deacetylation of NDUFS1 and/or Rieske subunit might have potential as a novel therapeutic approach for cardioprotection against ischemia/reperfusion.
Collapse
Affiliation(s)
- Ken Shinmura
- Division of Geriatric Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|