1
|
Ye Z, Liu R, Wang H, Zuo A, Jin C, Wang N, Sun H, Feng L, Yang H. Neuroprotective potential for mitigating ischemia-reperfusion-induced damage. Neural Regen Res 2025; 20:2199-2217. [PMID: 39104164 PMCID: PMC11759025 DOI: 10.4103/nrr.nrr-d-23-01985] [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: 02/24/2024] [Revised: 04/09/2024] [Accepted: 06/22/2024] [Indexed: 08/07/2024] Open
Abstract
Reperfusion following cerebral ischemia causes both structural and functional damage to brain tissue and could aggravate a patient's condition; this phenomenon is known as cerebral ischemia-reperfusion injury. Current studies have elucidated the neuroprotective role of the sirtuin protein family (Sirtuins) in modulating cerebral ischemia-reperfusion injury. However, the potential of utilizing it as a novel intervention target to influence the prognosis of cerebral ischemia-reperfusion injury requires additional exploration. In this review, the origin and research progress of Sirtuins are summarized, suggesting the involvement of Sirtuins in diverse mechanisms that affect cerebral ischemia-reperfusion injury, including inflammation, oxidative stress, blood-brain barrier damage, apoptosis, pyroptosis, and autophagy. The therapeutic avenues related to Sirtuins that may improve the prognosis of cerebral ischemia-reperfusion injury were also investigated by modulating Sirtuins expression and affecting representative pathways, such as nuclear factor-kappa B signaling, oxidative stress mediated by adenosine monophosphate-activated protein kinase, and the forkhead box O. This review also summarizes the potential of endogenous substances, such as RNA and hormones, drugs, dietary supplements, and emerging therapies that regulate Sirtuins expression. This review also reveals that regulating Sirtuins mitigates cerebral ischemia-reperfusion injury when combined with other risk factors. While Sirtuins show promise as a potential target for the treatment of cerebral ischemia-reperfusion injury, most recent studies are based on rodent models with circadian rhythms that are distinct from those of humans, potentially influencing the efficacy of Sirtuins-targeting drug therapies. Overall, this review provides new insights into the role of Sirtuins in the pathology and treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Zi Ye
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Runqing Liu
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hangxing Wang
- Division of Infectious Diseases, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aizhen Zuo
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Cen Jin
- School of Medical Imaging, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Nan Wang
- Division of Gastroenterology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huiqi Sun
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Luqian Feng
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hua Yang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
2
|
Ma YT, Li C, Shen Y, You WH, Han MX, Mu YF, Han FJ. Mechanisms of the JNK/p38 MAPK signaling pathway in drug resistance in ovarian cancer. Front Oncol 2025; 15:1533352. [PMID: 40352594 PMCID: PMC12063130 DOI: 10.3389/fonc.2025.1533352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal malignancy in the female reproductive system, and chemotherapy drug resistance is the main cause of treatment failure. The Mitogen-Activated Protein Kinases (MAPK) pathway plays a pivotal role in regulating cell proliferation, migration, and invasive capacity in response to extracellular stimuli. This review focuses on the mechanisms and therapeutic strategies related to the JNK/p38 MAPK signaling pathway in OC resistance. The JNK/p38 MAPK pathway plays a dual role in OC chemoresistance. This review examines its role in mediating OC treatment resistance by exploring the mechanisms of action of the JNK/p38 MAPK signaling pathway, particularly its involvement in several key biological processes, including apoptosis, autophagy, DNA damage response, the tumor microenvironment (TME), and drug efflux. Additionally, the review investigates the timing of activation of this pathway and its crosstalk with other signaling pathways such as PI3K/AKT and NF-κB. Targeting JNK/p38 MAPK signaling has shown promise in reversing chemoresistance, with several inhibitors and natural compounds demonstrating potential in preclinical studies. Regulating JNK/p38 MAPK may transform what was once a terminal obstacle into a manageable challenge for OC patients with chemotherapy resistance, ultimately improving survival and quality of life.
Collapse
Affiliation(s)
- Yu-Ting Ma
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Chan Li
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ying Shen
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Wan-Hui You
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ming-Xuan Han
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yi-Fan Mu
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Feng-Juan Han
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Pederson NJ, Diehl KL. DNA stimulates the deacetylase SIRT6 to mono-ADP-ribosylate proteins with histidine repeats. J Biol Chem 2025:108532. [PMID: 40280420 DOI: 10.1016/j.jbc.2025.108532] [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: 08/13/2024] [Revised: 03/19/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Sirtuins are the NAD+-dependent class III lysine deacylases (KDACs). Members of this family have been linked to longevity and a wide array of different diseases, motivating the pursuit of sirtuin modulator compounds. Sirtuin 6 (SIRT6) is a primarily nuclear KDAC that deacetylates histones to facilitate gene repression. In addition to this canonical post-translational modification (PTM) "eraser" function, SIRT6 can use NAD+ instead to "write" mono-ADP-ribosylation (mARylation) on target proteins. This enzymatic function has been primarily associated with SIRT6's role in the DNA damage response. This modification has been challenging to study because it is not clear under what precise cellular contexts it occurs, only a few substrates are known, and potential interference from other ADP-ribosyltransferases in cells, among other reasons. In this work, we used commercially available ADP-ribosylation detection reagents to investigate the mARylation activity of SIRT6 in a reconstituted system. We observed that SIRT6 is activated in its mARylation activity by binding to dsDNA ends. We further identified a surprising target motif within biochemical substrates of SIRT6, polyhistidine (polyHis) repeat tracts, that are present in several previously identified SIRT6 mARylation substrates. This work provides important context for SIRT6 mARylation activity, in contrast to its KDAC activity, and generates a list of new potential SIRT6 mARylation substrates based on the polyHis motif..
Collapse
|
4
|
Wang ZA, Markert J, Whedon SD, Abeywardana MY, Sheng X, Nam E, Lee K, Chen M, Waterbury A, Zhao Y, Farnung L, Cole PA. Structural and enzymatic plasticity of SIRT6 deacylase activity. J Biol Chem 2025; 301:108446. [PMID: 40147774 PMCID: PMC12051053 DOI: 10.1016/j.jbc.2025.108446] [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: 01/21/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
Sirtuin 6 (SIRT6) is an NAD-dependent protein deacylase that targets lysine residues in histones in the cell nucleus, where it helps maintain genome stability and links metabolism to epigenetic control. Dysregulation of SIRT6 is believed to be associated with aging and cancer, making it of pharmacological interest. In this study, we use cryo-EM and enzymology to explore SIRT6 preference and adaptability toward different nucleosomal substrates. We have visualized a trapped complex of SIRT6 in the process of deacylating H3K27, demonstrating how SIRT6 undergoes conformational changes to remove differently positioned histone marks. Additional biochemical studies further reveal the plasticity of SIRT6, which accommodates various metabolism-linked modifications, such as lysine lactylation and β-hydroxybutyrylation. To further understand the basis for substrate selectivity of SIRT6, we explore the effects of an established G60A enzyme mutation, proximal H3 modifications, and small-molecule modulators. These findings highlight the versatility of SIRT6 and provide key mechanistic insights into its molecular recognition.
Collapse
Affiliation(s)
- Zhipeng A Wang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States; Desai Sethi Urology Institute & Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jonathan Markert
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States
| | - Samuel D Whedon
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Maheeshi Yapa Abeywardana
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Xinlei Sheng
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Eunju Nam
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Kwangwoon Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Maggie Chen
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Amanda Waterbury
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States
| | - Yingming Zhao
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Lucas Farnung
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States.
| | - Philip A Cole
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States.
| |
Collapse
|
5
|
Guo B, Ma B, Li M, Li Y, Liang P, Han D, Yan X, Hu S. The nitration of SIRT6 aggravates neuronal damage during cerebral ischemia-reperfusion in rat. Nitric Oxide 2024; 153:26-40. [PMID: 39374645 DOI: 10.1016/j.niox.2024.10.004] [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: 06/25/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Ischemic stroke is a major cause of death and disability. The activation of neuronal nitric oxide synthase (nNOS) and the resulting production of nitric oxide (NO) via NMDA receptor-mediated calcium influx play an exacerbating role in cerebral ischemia reperfusion injury. The NO rapidly reacts with superoxide (O2-) to form peroxynitrite (ONOO-), a toxic molecule may modify proteins through tyrosine residue nitration, ultimately worsening neuronal damage. SIRT6 has been proven to be crucial in regulating cell proliferation, death, and aging in various pathological settings. We have previous reported that human SIRT6 tyrosine nitration decreased its intrinsic catalytic activity in vitro. However, the exact role of SIRT6 function in the process of cerebral ischemia reperfusion injury is not yet fully elucidated. Herein, we demonstrated that an increase in the nitration of SIRT6 led to reduce its enzymatic activity and aggravated hippocampal neuronal damage in a rat model of four-artery cerebral ischemia reperfusion. In addition, reducing SIRT6 nitration resulted in increase the activity of SIRT6, alleviating hippocampal neuronal damage. Moreover, SIRT6 nitration affected its downstream molecule activity such as PARP1 and GCN5, promoting the process of neuronal ischemic injury in rat hippocampus. Additionally, treatment with NMDA receptor antagonist MK801, or nNOS inhibitor 7-NI, and resveratrol (an antioxidant) diminished SIRT6 nitration and the catalytic activity of downstream molecules like PARP1 and GCN5, thereby reducing neuronal damage. Finally, in the biochemical regulation of SIRT6 activity, tyrosine 257 was essential for its activity and susceptibility to nitration. Replacing tyrosine 257 with phenylalanine in rat SIRT6 attenuated the death of SH-SY5Y neurocytes under oxygen-glucose deprivation (OGD) conditions. These results may offer further understanding of SIRT6 function in the pathogenesis of cerebral ischemic diseases.
Collapse
Affiliation(s)
- Bingnan Guo
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Bin Ma
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Department of Nuclear Medicine, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, 471000, China
| | - Ming Li
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Department of Emergency Medicine, The General Hospital of Xuzhou Mining Group, Xuzhou, Jiangsu, 221006, China
| | - Yuxin Li
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Pengchong Liang
- Department of Emergency Medicine, Central Hospital of Baoji City, Baoji, Shanxi, 721008, China
| | - Dong Han
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Xianliang Yan
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Department of Emergency Medicine, Suining People's Hospital, Xuzhou, Jiangsu, 221000, China.
| | - Shuqun Hu
- The Laboratory of Emergency Medicine, School of Second Clinical Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
6
|
Pederson NJ, Diehl KL. DNA stimulates SIRT6 to mono-ADP-ribosylate proteins within histidine repeats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606047. [PMID: 39211154 PMCID: PMC11361027 DOI: 10.1101/2024.07.31.606047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sirtuins are the NAD + -dependent class III lysine deacylases (KDACs). Members of this family have been linked to longevity and a wide array of different diseases, motivating the pursuit of sirtuin modulator compounds. Sirtuin 6 (SIRT6) is a primarily nuclear KDAC that deacetylates histones to facilitate gene repression. In addition to this canonical post-translational modification (PTM) "eraser" function, SIRT6 can use NAD + instead to "write" mono-ADP-ribosylation (mARylation) on target proteins. This enzymatic function has been primarily associated with SIRT6's role in the DNA damage response. This modification has been challenging to study because it is not clear under what precise cellular contexts it occurs, only a few substrates are known, and potential interference from other ADP-ribosyltransferases in cells, among other reasons. In this work, we used commercially available ADP-ribosylation detection reagents to investigate the mARylation activity of SIRT6 in a reconstituted system. We observed that SIRT6 is activated in its mARylation activity by binding to dsDNA ends. We further identified a surprising target motif within biochemical substrates of SIRT6, polyhistidine (polyHis) repeat tracts, that are present in several previously identified SIRT6 mARylation substrates and binding partners. This work provides important context for SIRT6 mARylation activity, in contrast to its KDAC activity, and proposes that SIRT6 is a histidine mARyltransferase enzyme.
Collapse
|
7
|
Yu L, Li Y, Song S, Zhang Y, Wang Y, Wang H, Yang Z, Wang Y. The dual role of sirtuins in cancer: biological functions and implications. Front Oncol 2024; 14:1384928. [PMID: 38947884 PMCID: PMC11211395 DOI: 10.3389/fonc.2024.1384928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024] Open
Abstract
Sirtuins are pivotal in orchestrating numerous cellular pathways, critically influencing cell metabolism, DNA repair, aging processes, and oxidative stress. In recent years, the involvement of sirtuins in tumor biology has garnered substantial attention, with a growing body of evidence underscoring their regulatory roles in various aberrant cellular processes within tumor environments. This article delves into the sirtuin family and its biological functions, shedding light on their dual roles-either as promoters or inhibitors-in various cancers including oral, breast, hepatocellular, lung, and gastric cancers. It further explores potential anti-tumor agents targeting sirtuins, unraveling the complex interplay between sirtuins, miRNAs, and chemotherapeutic drugs. The dual roles of sirtuins in cancer biology reflect the complexity of targeting these enzymes but also highlight the immense therapeutic potential. These advancements hold significant promise for enhancing clinical outcomes, marking a pivotal step forward in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Lu Yu
- Department of Respiratory, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanjiao Li
- Department of Pharmacy, Qionglai Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Yalin Zhang
- School of Medicine, University of Electronic Science and Technology of China, Center of Critical Care Medicine, Sichuan Academy of Medical Sciences, Chengdu, China
- Center of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yiping Wang
- Center of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science, Nanning, China
| | - Zhengteng Yang
- Department of Medicine, The First Affiliated Hospital of Guangxi University of Traditional Medicine, Nanning, China
| | - Yi Wang
- Center of Critical Care Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science, Nanning, China
| |
Collapse
|
8
|
Popov AA, Petruseva IO, Lavrik OI. Activity of DNA Repair Systems in the Cells of Long-Lived Rodents and Bats. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1014-1023. [PMID: 38981697 DOI: 10.1134/s0006297924060038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/15/2024] [Accepted: 04/03/2024] [Indexed: 07/11/2024]
Abstract
Damages of various origin accumulated in the genomic DNA can lead to the breach of genome stability, and are considered to be one of the main factors involved in cellular senescence. DNA repair systems in mammalian cells ensure effective damage removal and repair of the genome structure, therefore, activity of these systems is expected to be correlated with high maximum lifespan observed in the long-lived mammals. This review discusses current results of the studies focused on determination of the DNA repair system activity and investigation of the properties of its key regulatory proteins in the cells of long-lived rodents and bats. Based on the works discussed in the review, it could be concluded that the long-lived rodents and bats in general demonstrate high efficiency in functioning and regulation of DNA repair systems. Nevertheless, a number of questions around the study of DNA repair in the cells of long-lived rodents and bats remain poorly understood, answers to which could open up new avenues for further research.
Collapse
Affiliation(s)
- Aleksei A Popov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Irina O Petruseva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Olga I Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch Russian Academy of Sciences, Novosibirsk, 630090, Russia.
- Novosibirsk National Research State University, Novosibirsk, 630090, Russia
| |
Collapse
|
9
|
Divya KP, Kanwar N, Anuranjana PV, Kumar G, Beegum F, George KT, Kumar N, Nandakumar K, Kanwal A. SIRT6 in Regulation of Mitochondrial Damage and Associated Cardiac Dysfunctions: A Possible Therapeutic Target for CVDs. Cardiovasc Toxicol 2024; 24:598-621. [PMID: 38689163 DOI: 10.1007/s12012-024-09858-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Cardiovascular diseases (CVDs) can be described as a global health emergency imploring possible prevention strategies. Although the pathogenesis of CVDs has been extensively studied, the role of mitochondrial dysfunction in CVD development has yet to be investigated. Diabetic cardiomyopathy, ischemic-reperfusion injury, and heart failure are some of the CVDs resulting from mitochondrial dysfunction Recent evidence from the research states that any dysfunction of mitochondria has an impact on metabolic alteration, eventually causes the death of a healthy cell and therefore, progressively directing to the predisposition of disease. Cardiovascular research investigating the targets that both protect and treat mitochondrial damage will help reduce the risk and increase the quality of life of patients suffering from various CVDs. One such target, i.e., nuclear sirtuin SIRT6 is strongly associated with cardiac function. However, the link between mitochondrial dysfunction and SIRT6 concerning cardiovascular pathologies remains poorly understood. Although the Role of SIRT6 in skeletal muscles and cardiomyocytes through mitochondrial regulation has been well understood, its specific role in mitochondrial maintenance in cardiomyocytes is poorly determined. The review aims to explore the domain-specific function of SIRT6 in cardiomyocytes and is an effort to know how SIRT6, mitochondria, and CVDs are related.
Collapse
Affiliation(s)
- K P Divya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Navjot Kanwar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab, Technical University, Bathinda, Punjab, 151005, India
| | - P V Anuranjana
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
- School of Pharmacy, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology, National Institute of Pharmaceutical Educations and Research, Hajipur, Bihar, 844102, India
| | - K Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India.
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab, 151005, India.
| |
Collapse
|
10
|
Chen W, Mi C, Zhang Y, Yang Y, Huang W, Xu Z, Zhao J, Wang R, Wang M, Wan S, Wang X, Zhang H. Defective Homologous Recombination Repair By Up-Regulating Lnc-HZ10/Ahr Loop in Human Trophoblast Cells Induced Miscarriage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2207435. [PMID: 38286681 PMCID: PMC10987163 DOI: 10.1002/advs.202207435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/27/2023] [Indexed: 01/31/2024]
Abstract
Human trophoblast cells are crucial for healthy pregnancy. However, whether the defective homologous recombination (HR) repair of dsDNA break (DSB) in trophoblast cells may induce miscarriage is completely unknown. Moreover, the abundance of BRCA1 (a crucial protein for HR repair), its recruitment to DSB foci, and its epigenetic regulatory mechanisms, are also fully unexplored. In this work, it is identified that a novel lnc-HZ10, which is highly experssed in villous tissues of recurrent miscarriage (RM) vs their healthy control group, suppresses HR repair of DSB in trophoblast cell. Lnc-HZ10 and AhR (aryl hydrocarbon receptor) form a positive feedback loop. AhR acts as a transcription factor to promote lnc-HZ10 transcription. Meanwhile, lnc-HZ10 also increases AhR levels by suppressing its CUL4B-mediated ubiquitination degradation. Subsequently, AhR suppresses BRCA1 transcription; and lnc-HZ10 (mainly 1-447 nt) interacts with γ-H2AX; and thus, impairs its interactions with BRCA1. BPDE exposure may trigger this loop to suppress HR repair in trophoblast cells, possibly inducing miscarriage. Knockdown of murine Ahr efficiently recovers HR repair in placental tissues and alleviates miscarriage in a mouse miscarriage model. Therefore, it is suggested that AhR/lnc-HZ10/BRCA1 axis may be a promising target for alleviation of unexplained miscarriage.
Collapse
Affiliation(s)
- Weina Chen
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
- Key Laboratory of Environment and Female Reproductive HealthWest China School of Public Health & West China Fourth HospitalSichuan UniversityChengdu610041China
| | - Chenyang Mi
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Ying Zhang
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Yang Yang
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Wenxin Huang
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Zhongyan Xu
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Jingsong Zhao
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Rong Wang
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Manli Wang
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Shukun Wan
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Xiaoqing Wang
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| | - Huidong Zhang
- Research Center for Environment and Female Reproductive HealthThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhen518033China
| |
Collapse
|
11
|
Liu YL, Liu JY, Zhu XX, Wei JH, Mi SL, Liu SY, Li XL, Zhang WW, Zhao LL, Wang H, Xu DX, Gao L. Pubertal exposure to Microcystin-LR arrests spermatogonia proliferation by inducing DSB and inhibiting SIRT6 dependent DNA repair in vivo and in vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116191. [PMID: 38460408 DOI: 10.1016/j.ecoenv.2024.116191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
The reproduction toxicity of pubertal exposure to Microcystin-LR (MC-LR) and the underlying mechanism needs to be further investigated. In the current study, pubertal male ICR mice were intraperitoneally injected with 2 μg/kg MC-LR for four weeks. Pubertal exposure to MC-LR decreased epididymal sperm concentration and blocked spermatogonia proliferation. In-vitro studies found MC-LR inhibited cell proliferation of GC-1 cells and arrested cell cycle in G2/M phase. Mechanistically, MC-LR exposure evoked excessive reactive oxygen species (ROS) and induced DNA double-strand break in GC-1 cells. Besides, MC-LR inhibited DNA repair by reducing PolyADP-ribosylation (PARylation) activity of PARP1. Further study found MC-LR caused proteasomal degradation of SIRT6, a monoADP-ribosylation enzyme which is essential for PARP1 PARylation activity, due to destruction of SIRT6-USP10 interaction. Additionally, MG132 pretreatment alleviated MC-LR-induced SIRT6 degradation and promoted DNA repair, leading to the restoration of cell proliferation inhibition. Correspondingly, N-Acetylcysteine (NAC) pre-treatment mitigated the disturbed SIRT6-USP10 interaction and SIRT6 degradation, causing recovered DNA repair and subsequently restoration of cell proliferation inhibition in MC-LR treated GC-1 cells. Together, pubertal exposure to MC-LR induced spermatogonia cell cycle arrest and sperm count reduction by oxidative DNA damage and simultaneous SIRT6-mediated DNA repair failing. This study reports the effect of pubertal exposure to MC-LR on spermatogenesis and complex mechanism how MC-LR induces spermatogonia cell proliferation inhibition.
Collapse
Affiliation(s)
- Yu-Lin Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Jia-Yu Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Xin-Xin Zhu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Jian-Hua Wei
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Shuang-Ling Mi
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Su-Ya Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Xiu-Liang Li
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Wei-Wei Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Ling-Li Zhao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Hua Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - De-Xiang Xu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China.
| | - Lan Gao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, China; Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China.
| |
Collapse
|
12
|
Wang TH, Shen YW, Chen HY, Chen CC, Lin NC, Shih YH, Hsia SM, Chiu KC, Shieh TM. Arecoline Induces ROS Accumulation, Transcription of Proinflammatory Factors, and Expression of KRT6 in Oral Epithelial Cells. Biomedicines 2024; 12:412. [PMID: 38398015 PMCID: PMC10887121 DOI: 10.3390/biomedicines12020412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Areca nut is a major contributor to the high prevalence of oral cancer in Asia. The precise mechanisms by which areca nut stimulates mucosal cells and contributes to the progression of oral cancer urgently require clarification. The current study aimed to assess the effects of arecoline on the normal human gingival epithelium cell line S-G. Cell viability, levels of reactive oxygen species (ROS), protein expression, cellular morphology, and gene expression were evaluated using the MTT test, flow cytometry, Western blot analysis, optical or confocal microscopy, and RT-qPCR. Keratin (KRT6) analysis involved matched normal and cancer tissues from clinical head and neck specimens. The results demonstrated that 12.5 µg/mL of arecoline induced ROS production, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) mRNA expression in S-G cells. This activation of the MAPK/ERK pathway increased KRT6 expression while limiting cell migration. In head and neck cancer tissues, KRT6B gene expression exceeded that of normal tissues. This study confirms that arecoline induces ROS accumulation in normal cells, leading to the secretion of proinflammatory factors and KRT6 expression. This impedes oral mucosal healing, thereby promoting the progression of oral cancer.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Biobank, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yen-Wen Shen
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
| | - Hsin-Ying Chen
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
| | - Chih-Chieh Chen
- Department of Sports Medicine, China Medical University, Taichung 404328, Taiwan
| | - Nan-Chin Lin
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
- Department of Oral and Maxillofacial Surgery, Show Chwan Memorial Hospital, Changhua 505029, Taiwan
- Department of Oral and Maxillofacial Surgery, Changhua Christian Hospital, Changhua 500011, Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Kuo-Chou Chiu
- Division of General Dentistry, Taichung Armed Forces General Hospital, Taichung 411228, Taiwan
| | - Tzong-Ming Shieh
- School of Dentistry, China Medical University, Taichung 404328, Taiwan
| |
Collapse
|
13
|
Affandi T, Haas A, Ohm AM, Wright GM, Black JC, Reyland ME. PKCδ Regulates Chromatin Remodeling and DNA Repair through SIRT6. Mol Cancer Res 2024; 22:181-196. [PMID: 37889141 PMCID: PMC10872792 DOI: 10.1158/1541-7786.mcr-23-0493] [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: 06/26/2023] [Revised: 09/07/2023] [Accepted: 10/25/2023] [Indexed: 10/28/2023]
Abstract
Irradiation (IR) is a highly effective cancer therapy; however, IR damage to tumor-adjacent healthy tissues can result in significant comorbidities and potentially limit the course of therapy. We have previously shown that protein kinase C delta (PKCδ) is required for IR-induced apoptosis and that inhibition of PKCδ activity provides radioprotection in vivo. Here we show that PKCδ regulates histone modification, chromatin accessibility, and double-stranded break (DSB) repair through a mechanism that requires Sirtuin 6 (SIRT6). Overexpression of PKCδ promotes genomic instability and increases DNA damage and apoptosis. Conversely, depletion of PKCδ increases DNA repair via nonhomologous end joining (NHEJ) and homologous recombination (HR) as evidenced by increased formation of DNA damage foci, increased expression of DNA repair proteins, and increased repair of NHEJ and HR fluorescent reporter constructs. Nuclease sensitivity indicates that PKCδ depletion is associated with more open chromatin, while overexpression of PKCδ reduces chromatin accessibility. Epiproteome analysis reveals increased chromatin associated H3K36me2 in PKCδ-depleted cells which is accompanied by chromatin disassociation of KDM2A. We identify SIRT6 as a downstream mediator of PKCδ. PKCδ-depleted cells have increased SIRT6 expression, and depletion of SIRT6 reverses changes in chromatin accessibility, histone modification and DSB repair in PKCδ-depleted cells. Furthermore, depletion of SIRT6 reverses radioprotection in PKCδ-depleted cells. Our studies describe a novel pathway whereby PKCδ orchestrates SIRT6-dependent changes in chromatin accessibility to regulate DNA repair, and define a mechanism for regulation of radiation-induced apoptosis by PKCδ. IMPLICATIONS PKCδ controls sensitivity to irradiation by regulating DNA repair.
Collapse
Affiliation(s)
- Trisiani Affandi
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ami Haas
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angela M. Ohm
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Gregory M. Wright
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joshua C. Black
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mary E. Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
14
|
Wei W, Li T, Chen J, Fan Z, Gao F, Yu Z, Jiang Y. SIRT3/6: an amazing challenge and opportunity in the fight against fibrosis and aging. Cell Mol Life Sci 2024; 81:69. [PMID: 38294557 PMCID: PMC10830597 DOI: 10.1007/s00018-023-05093-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 02/01/2024]
Abstract
Fibrosis is a typical aging-related pathological process involving almost all organs, including the heart, kidney, liver, lung, and skin. Fibrogenesis is a highly orchestrated process defined by sequences of cellular response and molecular signals mechanisms underlying the disease. In pathophysiologic conditions associated with organ fibrosis, a variety of injurious stimuli such as metabolic disorders, epigenetic changes, and aging may induce the progression of fibrosis. Sirtuins protein is a kind of deacetylase which can regulate cell metabolism and participate in a variety of cell physiological functions. In this review, we outline our current understanding of common principles of fibrogenic mechanisms and the functional role of SIRT3/6 in aging-related fibrosis. In addition, sequences of novel protective strategies have been identified directly or indirectly according to these mechanisms. Here, we highlight the role and biological function of SIRT3/6 focus on aging fibrosis, as well as their inhibitors and activators as novel preventative or therapeutic interventions for aging-related tissue fibrosis.
Collapse
Affiliation(s)
- Wenxin Wei
- School of Queen Mary, Nanchang University, Nanchang, 330031, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jinlong Chen
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China
| | - Zhen Fan
- The Hospital Affiliated to Shanxi University of Chinese Medicine, Xianyang, 712000, China.
| | - Feng Gao
- Shanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Zhibiao Yu
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China
| | - Yihao Jiang
- School of Chemistry and Chemical Engineering, Nangchang University, 999 Xuefu Rd, Nanchang, 330031, China.
| |
Collapse
|
15
|
Waters KL, Spratt DE. New Discoveries on Protein Recruitment and Regulation during the Early Stages of the DNA Damage Response Pathways. Int J Mol Sci 2024; 25:1676. [PMID: 38338953 PMCID: PMC10855619 DOI: 10.3390/ijms25031676] [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: 12/19/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Maintaining genomic stability and properly repairing damaged DNA is essential to staying healthy and preserving cellular homeostasis. The five major pathways involved in repairing eukaryotic DNA include base excision repair (BER), nucleotide excision repair (NER), mismatch repair (MMR), non-homologous end joining (NHEJ), and homologous recombination (HR). When these pathways do not properly repair damaged DNA, genomic stability is compromised and can contribute to diseases such as cancer. It is essential that the causes of DNA damage and the consequent repair pathways are fully understood, yet the initial recruitment and regulation of DNA damage response proteins remains unclear. In this review, the causes of DNA damage, the various mechanisms of DNA damage repair, and the current research regarding the early steps of each major pathway were investigated.
Collapse
Affiliation(s)
| | - Donald E. Spratt
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, 950 Main St., Worcester, MA 01610, USA;
| |
Collapse
|
16
|
Todorova MN, Savova MS, Mihaylova LV, Georgiev MI. Icariin Improves Stress Resistance and Extends Lifespan in Caenorhabditis elegans through hsf-1 and daf-2-Driven Hormesis. Int J Mol Sci 2023; 25:352. [PMID: 38203522 PMCID: PMC10778813 DOI: 10.3390/ijms25010352] [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: 11/27/2023] [Revised: 12/17/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Aging presents an increasingly significant challenge globally, driven by the growing proportion of individuals aged 60 and older. Currently, there is substantial research interest in pro-longevity interventions that target pivotal signaling pathways, aiming not only to extend lifespan but also to enhance healthspan. One particularly promising approach involves inducing a hormetic response through the utilization of natural compounds defined as hormetins. Various studies have introduced the flavonoid icariin as beneficial for age-related diseases such as cardiovascular and neurodegenerative conditions. To validate its potential pro-longevity properties, we employed Caenorhabditis elegans as an experimental platform. The accumulated results suggest that icariin extends the lifespan of C. elegans through modulation of the DAF-2, corresponding to the insulin/IGF-1 signaling pathway in humans. Additionally, we identified increased resistance to heat and oxidative stress, modulation of lipid metabolism, improved late-life healthspan, and an extended lifespan upon icariin treatment. Consequently, a model mechanism of action was provided for icariin that involves the modulation of various players within the stress-response network. Collectively, the obtained data reveal that icariin is a potential hormetic agent with geroprotective properties that merits future developments.
Collapse
Affiliation(s)
- Monika N. Todorova
- Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000 Plovdiv, Bulgaria; (M.N.T.); (M.S.S.); (L.V.M.)
| | - Martina S. Savova
- Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000 Plovdiv, Bulgaria; (M.N.T.); (M.S.S.); (L.V.M.)
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
| | - Liliya V. Mihaylova
- Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000 Plovdiv, Bulgaria; (M.N.T.); (M.S.S.); (L.V.M.)
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
| | - Milen I. Georgiev
- Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000 Plovdiv, Bulgaria; (M.N.T.); (M.S.S.); (L.V.M.)
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
| |
Collapse
|
17
|
Kang W, Hamza A, Curry AM, Korade E, Donu D, Cen Y. Activation of SIRT6 Deacetylation by DNA Strand Breaks. ACS OMEGA 2023; 8:41310-41320. [PMID: 37970049 PMCID: PMC10633859 DOI: 10.1021/acsomega.3c04859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/05/2023] [Indexed: 11/17/2023]
Abstract
SIRT6 is an emerging regulator of longevity. Overexpression of SIRT6 extends the lifespan of mice. Conversely, SIRT6 knockout mice demonstrate severe metabolic defects and a shortened lifespan. The discrepancy between SIRT6's weak in vitro activity and robust in vivo activity has led to the hypothesis that this enzyme can be activated in response to DNA damage in cells. Here, we demonstrate that the deacetylase activity of SIRT6 can be stimulated by DNA strand breaks for synthetic peptide and histone substrates. The mechanism of activation is further explored by using an integrative chemical biology approach. SIRT6 can be preferentially activated by DNA lesions harboring a 5'-phosphate. The N- and C-termini of SIRT6 are strictly required for DNA break-induced activation. Additionally, the defatty-acylase activity of SIRT6 is also sensitive to DNA breaks, although the physiological significance needs further investigation. Collectively, our study sheds important light on the cellular regulation of diverse SIRT6 activities and suggests possible strategies for effective SIRT6 activation.
Collapse
Affiliation(s)
- Wenjia Kang
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298-0540, United States
| | - Abu Hamza
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298-0540, United States
| | - Alyson M. Curry
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298-0540, United States
| | - Evan Korade
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298-0540, United States
| | - Dickson Donu
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298-0540, United States
| | - Yana Cen
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298-0540, United States
- Institute
for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23298-0133, United States
| |
Collapse
|
18
|
Cheng D, Zheng B, Sheng Y, Zeng Z, Mo Z. The Roles of Autophagy in the Genesis and Development of Polycystic Ovary Syndrome. Reprod Sci 2023; 30:2920-2931. [PMID: 37204635 DOI: 10.1007/s43032-023-01255-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/29/2023] [Indexed: 05/20/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common and frequent disease and always leads endocrine and metabolic disorder among women in reproductive age. Ovary is the main organ involved in polycystic ovary syndrome, and its function impairment will lead to reproductive dysfunction. Some recent studies have demonstrated that autophagy plays an important role in the pathogenesis of PCOS, and there are many different mechanisms that affect autophagy and the occurrence of PCOS, and they provide a new direction for us to predict the mechanism of PCOS. In this review, we discuss the role of autophagy in different ovarian cells: granulosa cells, oocytes, and theca cells, and introduce the important role that they play in the progress of PCOS. The main purpose of this review is to provide the research background and some relevant suggestions for our future work in autophagy and help us better explore the pathogenesis and autophagy mechanisms of PCOS. Furthermore, it will help us gain a new insight of the pathophysiology and treatment of PCOS.
Collapse
Affiliation(s)
- Di Cheng
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guangxi, 541199, Guilin, China
- Joint Laboratory of Chronic Disease Prevention and Research, Guilin Medical University, Hunan Mingshun Pharmaceutical Co., Ltd, Shaodong, Hunan, 422800, Guilin, China
| | - Biao Zheng
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guangxi, 541199, Guilin, China
- Joint Laboratory of Chronic Disease Prevention and Research, Guilin Medical University, Hunan Mingshun Pharmaceutical Co., Ltd, Shaodong, Hunan, 422800, Guilin, China
| | - Ying Sheng
- Department of Obstetrics, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Zhaoming Zeng
- Joint Laboratory of Chronic Disease Prevention and Research, Guilin Medical University, Hunan Mingshun Pharmaceutical Co., Ltd, Shaodong, Hunan, 422800, Guilin, China.
| | - Zhongcheng Mo
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guangxi, 541199, Guilin, China.
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China.
| |
Collapse
|
19
|
Zhao K, Zheng M, Su Z, Ghosh S, Zhang C, Zhong W, Ho JWK, Jin G, Zhou Z. MOF-mediated acetylation of SIRT6 disrupts SIRT6-FOXA2 interaction and represses SIRT6 tumor-suppressive function by upregulating ZEB2 in NSCLC. Cell Rep 2023; 42:112939. [PMID: 37566546 DOI: 10.1016/j.celrep.2023.112939] [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: 10/10/2022] [Revised: 06/05/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Mammalian sirtuin 6 (SIRT6) regulates a spectrum of vital biological processes and has long been implicated in the progression of cancer. However, the mechanisms underlying the regulation of SIRT6 in tumorigenesis remain elusive. Here, we report that the tumor-suppressive function of SIRT6 in non-small cell lung cancer (NSCLC) is regulated by acetylation. Specifically, males absent on the first (MOF) acetylates SIRT6 at K128, K160, and K267, resulting in a decreased deacetylase activity of SIRT6 and attenuated SIRT6 tumor-suppressive function in NSCLC. Mechanistically, MOF-mediated SIRT6 acetylation hinders the interaction between SIRT6 and transcriptional factor FOXA2, which in turn leads to the transcriptional activation of ZEB2, thus promoting NSCLC progression. Collectively, these data indicate an acetylation-dependent mechanism that modulates SIRT6 tumor-suppressive function in NSCLC. Our findings suggest that the MOF-SIRT6-ZEB2 axis may represent a promising therapeutic target for the management of NSCLC.
Collapse
Affiliation(s)
- Kaiqiang Zhao
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P.R. China; School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China
| | - Mingyue Zheng
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P.R. China
| | - Zezhuo Su
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, P.R. China; Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, P.R. China
| | - Shrestha Ghosh
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, P.R. China; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, P.R. China
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, P.R. China
| | - Joshua Wing Kei Ho
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, P.R. China; Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, P.R. China
| | - Guoxiang Jin
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P.R. China.
| | - Zhongjun Zhou
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, P.R. China; School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China; Reproductive Medical Center, The University of Hong Kong Shenzhen Hospital, Shenzhen, P.R. China.
| |
Collapse
|
20
|
Deng Y, Adam V, Nepovimova E, Heger Z, Valko M, Wu Q, Wei W, Kuca K. c-Jun N-terminal kinase signaling in cellular senescence. Arch Toxicol 2023; 97:2089-2109. [PMID: 37335314 DOI: 10.1007/s00204-023-03540-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Cellular senescence leads to decreased tissue regeneration and inflammation and is associated with diabetes, neurodegenerative diseases, and tumorigenesis. However, the mechanisms of cellular senescence are not fully understood. Emerging evidence has indicated that c-Jun N-terminal kinase (JNK) signaling is involved in the regulation of cellular senescence. JNK can downregulate hypoxia inducible factor-1α to accelerate hypoxia-induced neuronal cell senescence. The activation of JNK inhibits mTOR activity and triggers autophagy, which promotes cellular senescence. JNK can upregulate the expression of p53 and Bcl-2 and accelerates cancer cell senescence; however, this signaling also mediates the expression of amphiregulin and PD-LI to achieve cancer cell immune evasion and prevents their senescence. The activation of JNK further triggers forkhead box O expression and its target gene Jafrac1 to extend the lifespan of Drosophila. JNK can also upregulate the expression of DNA repair protein poly ADP-ribose polymerase 1 and heat shock protein to delay cellular senescence. This review discusses recent advances in understanding the function of JNK signaling in cellular senescence and includes a comprehensive analysis of the molecular mechanisms underlying JNK-mediated senescence evasion and oncogene-induced cellular senescence. We also summarize the research progress in anti-aging agents that target JNK signaling. This study will contribute to a better understanding of the molecular targets of cellular senescence and provides insights into anti-aging, which may be used to develop drugs for the treatment of aging-related diseases.
Collapse
Affiliation(s)
- Ying Deng
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
| | - Wei Wei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain.
| |
Collapse
|
21
|
Wu T, Qu Y, Xu S, Wang Y, Liu X, Ma D. SIRT6: A potential therapeutic target for diabetic cardiomyopathy. FASEB J 2023; 37:e23099. [PMID: 37462453 DOI: 10.1096/fj.202301012r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
The abnormal lipid metabolism in diabetic cardiomyopathy can cause myocardial mitochondrial dysfunction, lipotoxicity, abnormal death of myocardial cells, and myocardial remodeling. Mitochondrial homeostasis and normal lipid metabolism can effectively slow down the development of diabetic cardiomyopathy. Recent studies have shown that SIRT6 may play an important role in the pathological changes of diabetic cardiomyopathy such as myocardial cell death, myocardial hypertrophy, and myocardial fibrosis by regulating mitochondrial oxidative stress and glucose and lipid metabolism. Therefore, understanding the function of SIRT6 and its role in the pathogenesis of diabetic cardiomyopathy is of great significance for exploring and developing new targets and drugs for the treatment of diabetic cardiomyopathy. This article reviews the latest findings of SIRT6 in the pathogenesis of diabetic cardiomyopathy, focusing on the regulation of mitochondria and lipid metabolism by SIRT6 to explore potential clinical treatments.
Collapse
Affiliation(s)
- Tao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiwei Qu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shengjie Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yong Wang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Xue Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dufang Ma
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
22
|
Wu K, Wang Y, Liu R, Wang H, Rui T. The role of mammalian Sirtuin 6 in cardiovascular diseases and diabetes mellitus. Front Physiol 2023; 14:1207133. [PMID: 37497437 PMCID: PMC10366693 DOI: 10.3389/fphys.2023.1207133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 07/28/2023] Open
Abstract
Cardiovascular diseases are severe diseases posing threat to human health because of their high morbidity and mortality worldwide. The incidence of diabetes mellitus is also increasing rapidly. Various signaling molecules are involved in the pathogenesis of cardiovascular diseases and diabetes. Sirtuin 6 (Sirt6), which is a class III histone deacetylase, has attracted numerous attentions since its discovery. Sirt6 enjoys a unique structure, important biological functions, and is involved in multiple cellular processes such as stress response, mitochondrial biogenesis, transcription, insulin resistance, inflammatory response, chromatin silencing, and apoptosis. Sirt6 also plays significant roles in regulating several cardiovascular diseases including atherosclerosis, coronary heart disease, as well as cardiac remodeling, bringing Sirt6 into the focus of clinical interests. In this review, we examine the recent advances in understanding the mechanistic working through which Sirt6 alters the course of lethal cardiovascular diseases and diabetes mellitus.
Collapse
|
23
|
Affandi T, Haas A, Ohm AM, Wright GM, Black JC, Reyland ME. PKCδ regulates chromatin remodeling and DNA repair through SIRT6. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.541991. [PMID: 37292592 PMCID: PMC10245827 DOI: 10.1101/2023.05.24.541991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Protein kinase C delta (PKCδ) is a ubiquitous kinase whose function is defined in part by localization to specific cellular compartments. Nuclear PKCδ is both necessary and sufficient for IR-induced apoptosis, while inhibition of PKCδ activity provides radioprotection in vivo. How nuclear PKCδ regulates DNA-damage induced cell death is poorly understood. Here we show that PKCδ regulates histone modification, chromatin accessibility, and double stranded break (DSB) repair through a mechanism that requires SIRT6. Overexpression of PKCδ promotes genomic instability and increases DNA damage and apoptosis. Conversely, depletion of PKCδ increases DNA repair via non-homologous end joining (NHEJ) and homologous recombination (HR) as evidenced by more rapid formation of NHEJ (DNA-PK) and HR (Rad51) DNA damage foci, increased expression of repair proteins, and increased repair of NHEJ and HR fluorescent reporter constructs. Nuclease sensitivity indicates that PKCδ depletion is associated with more open chromatin, while overexpression of PKCδ reduces chromatin accessibility. Epiproteome analysis revealed that PKCδ depletion increases chromatin associated H3K36me2, and reduces ribosylation of KDM2A and chromatin bound KDM2A. We identify SIRT6 as a downstream mediator of PKCδ. PKCδ-depleted cells have increased expression of SIRT6, and depletion of SIRT6 reverses the changes in chromatin accessibility, histone modification and NHEJ and HR DNA repair seen with PKCδ-depletion. Furthermore, depletion of SIRT6 reverses radioprotection in PKCδ-depleted cells. Our studies describe a novel pathway whereby PKCδ orchestrates SIRT6-dependent changes in chromatin accessibility to increase DNA repair, and define a mechanism for regulation of radiation-induced apoptosis by PKCδ.
Collapse
Affiliation(s)
- Trisiani Affandi
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ami Haas
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angela M. Ohm
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Gregory M. Wright
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joshua C. Black
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mary E. Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
24
|
Kim D, Orr MJ, Kwong AJ, Deibler KK, Munshi HH, Bridges CS, Chen TJ, Zhang X, Lacorazza HD, Scheidt KA. Rational Design of Highly Potent and Selective Covalent MAP2K7 Inhibitors. ACS Med Chem Lett 2023; 14:606-613. [PMID: 37197477 PMCID: PMC10184151 DOI: 10.1021/acsmedchemlett.3c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/10/2023] [Indexed: 05/19/2023] Open
Abstract
The mitogen-activated protein kinase signaling cascade is conserved across eukaryotes, where it plays a critical role in the regulation of activities including proliferation, differentiation, and stress responses. This pathway propagates external stimuli through a series of phosphorylation events, which allows external signals to influence metabolic and transcriptional activities. Within the cascade, MEK, or MAP2K, enzymes occupy a molecular crossroads immediately upstream to significant signal divergence and cross-talk. One such kinase, MAP2K7, also known as MEK7 and MKK7, is a protein of great interest in the molecular pathophysiology underlying pediatric T cell acute lymphoblastic leukemia (T-ALL). Herein, we describe the rational design, synthesis, evaluation, and optimization of a novel class of irreversible MAP2K7 inhibitors. With a streamlined one-pot synthesis, favorable in vitro potency and selectivity, and promising cellular activity, this novel class of compounds wields promise as a powerful tool in the study of pediatric T-ALL.
Collapse
Affiliation(s)
- Dalton
R. Kim
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Meghan J. Orr
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Ada J. Kwong
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Kristine K. Deibler
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Hasan H. Munshi
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Cory Seth Bridges
- Department
of Pathology & Immunology, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Taylor Jie Chen
- Department
of Pathology & Immunology, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Xiaoyu Zhang
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, Evanston, Illinois 60208, United
States
| | - H. Daniel Lacorazza
- Department
of Pathology & Immunology, Baylor College
of Medicine, Houston, Texas 77030, United States
| | - Karl A. Scheidt
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, Evanston, Illinois 60208, United
States
- Department
of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
25
|
Carlsson R, Enström A, Paul G. Molecular Regulation of the Response of Brain Pericytes to Hypoxia. Int J Mol Sci 2023; 24:5671. [PMID: 36982744 PMCID: PMC10053233 DOI: 10.3390/ijms24065671] [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: 02/19/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
The brain needs sufficient oxygen in order to function normally. This is achieved by a large vascular capillary network ensuring that oxygen supply meets the changing demand of the brain tissue, especially in situations of hypoxia. Brain capillaries are formed by endothelial cells and perivascular pericytes, whereby pericytes in the brain have a particularly high 1:1 ratio to endothelial cells. Pericytes not only have a key location at the blood/brain interface, they also have multiple functions, for example, they maintain blood-brain barrier integrity, play an important role in angiogenesis and have large secretory abilities. This review is specifically focused on both the cellular and the molecular responses of brain pericytes to hypoxia. We discuss the immediate early molecular responses in pericytes, highlighting four transcription factors involved in regulating the majority of transcripts that change between hypoxic and normoxic pericytes and their potential functions. Whilst many hypoxic responses are controlled by hypoxia-inducible factors (HIF), we specifically focus on the role and functional implications of the regulator of G-protein signaling 5 (RGS5) in pericytes, a hypoxia-sensing protein that is regulated independently of HIF. Finally, we describe potential molecular targets of RGS5 in pericytes. These molecular events together contribute to the pericyte response to hypoxia, regulating survival, metabolism, inflammation and induction of angiogenesis.
Collapse
Affiliation(s)
- Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
- Department of Neurology, Scania University Hospital, 22185 Lund, Sweden
| |
Collapse
|
26
|
Abstract
The HER2+ subtype of human breast cancer is associated with the malignant transformation of luminal ductal cells of the mammary epithelium. The sequence analysis of tumor DNA identifies loss of function mutations and deletions of the MAP2K4 and MAP2K7 genes that encode direct activators of the JUN NH2-terminal kinase (JNK). We report that in vitro studies of human mammary epithelial cells with CRISPR-induced mutations in the MAPK and MAP2K components of the JNK pathway caused no change in growth in 2D culture, but these mutations promoted epithelial cell proliferation in 3D culture. Analysis of gene expression signatures in 3D culture demonstrated similar changes caused by HER2 activation and JNK pathway loss. The mechanism of signal transduction cross-talk may be mediated, in part, by JNK-suppressed expression of integrin α6β4 that binds HER2 and amplifies HER2 signaling. These data suggest that HER2 activation and JNK pathway loss may synergize to promote breast cancer. To test this hypothesis, we performed in vivo studies using a mouse model of HER2+ breast cancer with Cre/loxP-mediated ablation of genes encoding JNK (Mapk8 and Mapk9) and the MAP2K (Map2k4 and Map2k7) that activate JNK in mammary epithelial cells. Kaplan-Meier analysis of tumor development demonstrated that JNK pathway deficiency promotes HER2+-driven breast cancer. Collectively, these data identify JNK pathway genes as potential suppressors for HER2+ breast cancer.
Collapse
|
27
|
Shah ZA, Nouroz F, Ejaz S, Tayyeb A. An Insight into the Role of E2F1 in Breast Cancer Progression, Drug Resistance, and Metastasis. Curr Mol Med 2023; 23:365-376. [PMID: 35260053 DOI: 10.2174/1566524022666220308095834] [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: 05/15/2021] [Revised: 11/25/2021] [Accepted: 12/07/2021] [Indexed: 11/22/2022]
Abstract
AIMS This study aimed to investigate the role of E2F1 in breast cancer biology. BACKGROUND Expression of E2F1, a transcription factor of many oncogenes and tumor suppressor genes, is lowered in several malignancies, including breast carcinoma. OBJECTIVES In the present study, we analyzed the status of E2F1 expression in association with diverse attributes of breast malignancy and its impact on cancer progression. METHODS For this purpose, we used various freely available online applications for gene enrichment, expression, and methylation analysis to extract mutation-based E2F1 map, to measure E2F1 drug sensitivity, and to determine E2F1 association with DNA damage response proteins. RESULTS Results revealed tissue-specific regulatory behavior of E2F1. Moreover, the key role of E2F1 in the promotion of metastasis, stem cell-mediated carcinogenesis, estrogen-mediated cell proliferation, and cellular defense system, has therefore highlighted it as a metaplastic marker and hot member of key resistome pathways. CONCLUSION The information thus generated can be employed for future implications in devising rational therapeutic strategies. Moreover, this study has provided a more detailed insight into the diagnostic and prognostic potential of E2F1.
Collapse
Affiliation(s)
- Zafar Abbas Shah
- Department of Bioinformatics, Hazara University Mansehra, Mansehra, Pakistan
| | - Faisal Nouroz
- Department of Bioinformatics, Hazara University Mansehra, Mansehra, Pakistan
| | - Samina Ejaz
- Department of Biochemistry, Institute of Biochemistry, Biotechnology and Bioinformatics (IBBB), The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Asima Tayyeb
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
28
|
Guo Z, Li P, Ge J, Li H. SIRT6 in Aging, Metabolism, Inflammation and Cardiovascular Diseases. Aging Dis 2022; 13:1787-1822. [PMID: 36465178 PMCID: PMC9662279 DOI: 10.14336/ad.2022.0413] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/13/2022] [Indexed: 07/28/2023] Open
Abstract
As an important NAD+-dependent enzyme, SIRT6 has received significant attention since its discovery. In view of observations that SIRT6-deficient animals exhibit genomic instability and metabolic disorders and undergo early death, SIRT6 has long been considered a protein of longevity. Recently, growing evidence has demonstrated that SIRT6 functions as a deacetylase, mono-ADP-ribosyltransferase and long fatty deacylase and participates in a variety of cellular signaling pathways from DNA damage repair in the early stage to disease progression. In this review, we elaborate on the specific substrates and molecular mechanisms of SIRT6 in various physiological and pathological processes in detail, emphasizing its links to aging (genomic damage, telomere integrity, DNA repair), metabolism (glycolysis, gluconeogenesis, insulin secretion and lipid synthesis, lipolysis, thermogenesis), inflammation and cardiovascular diseases (atherosclerosis, cardiac hypertrophy, heart failure, ischemia-reperfusion injury). In addition, the most recent advances regarding SIRT6 modulators (agonists and inhibitors) as potential therapeutic agents for SIRT6-mediated diseases are reviewed.
Collapse
Affiliation(s)
- Zhenyang Guo
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| | - Peng Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hua Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Zhao P, Malik S. The phosphorylation to acetylation/methylation cascade in transcriptional regulation: how kinases regulate transcriptional activities of DNA/histone-modifying enzymes. Cell Biosci 2022; 12:83. [PMID: 35659740 PMCID: PMC9164400 DOI: 10.1186/s13578-022-00821-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022] Open
Abstract
Transcription factors directly regulate gene expression by recognizing and binding to specific DNA sequences, involving the dynamic alterations of chromatin structure and the formation of a complex with different kinds of cofactors, like DNA/histone modifying-enzymes, chromatin remodeling factors, and cell cycle factors. Despite the significance of transcription factors, it remains unclear to determine how these cofactors are regulated to cooperate with transcription factors, especially DNA/histone modifying-enzymes. It has been known that DNA/histone modifying-enzymes are regulated by post-translational modifications. And the most common and important modification is phosphorylation. Even though various DNA/histone modifying-enzymes have been classified and partly explained how phosphorylated sites of these enzymes function characteristically in recent studies. It still needs to find out the relationship between phosphorylation of these enzymes and the diseases-associated transcriptional regulation. Here this review describes how phosphorylation affects the transcription activity of these enzymes and other functions, including protein stability, subcellular localization, binding to chromatin, and interaction with other proteins.
Collapse
|
30
|
Simon M, Yang J, Gigas J, Earley EJ, Hillpot E, Zhang L, Zagorulya M, Tombline G, Gilbert M, Yuen SL, Pope A, Van Meter M, Emmrich S, Firsanov D, Athreya A, Biashad SA, Han J, Ryu S, Tare A, Zhu Y, Hudgins A, Atzmon G, Barzilai N, Wolfe A, Moody K, Garcia BA, Thomas DD, Robbins PD, Vijg J, Seluanov A, Suh Y, Gorbunova V. A rare human centenarian variant of SIRT6 enhances genome stability and interaction with Lamin A. EMBO J 2022; 41:e110393. [PMID: 36215696 PMCID: PMC9627671 DOI: 10.15252/embj.2021110393] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 02/02/2023] Open
Abstract
Sirtuin 6 (SIRT6) is a deacylase and mono-ADP ribosyl transferase (mADPr) enzyme involved in multiple cellular pathways implicated in aging and metabolism regulation. Targeted sequencing of SIRT6 locus in a population of 450 Ashkenazi Jewish (AJ) centenarians and 550 AJ individuals without a family history of exceptional longevity identified enrichment of a SIRT6 allele containing two linked substitutions (N308K/A313S) in centenarians compared with AJ control individuals. Characterization of this SIRT6 allele (centSIRT6) demonstrated it to be a stronger suppressor of LINE1 retrotransposons, confer enhanced stimulation of DNA double-strand break repair, and more robustly kill cancer cells compared with wild-type SIRT6. Surprisingly, centSIRT6 displayed weaker deacetylase activity, but stronger mADPr activity, over a range of NAD+ concentrations and substrates. Additionally, centSIRT6 displayed a stronger interaction with Lamin A/C (LMNA), which was correlated with enhanced ribosylation of LMNA. Our results suggest that enhanced SIRT6 function contributes to human longevity by improving genome maintenance via increased mADPr activity and enhanced interaction with LMNA.
Collapse
Affiliation(s)
- Matthew Simon
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Jiping Yang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Jonathan Gigas
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Eric J Earley
- Biostatistics and Epidemiology, RTI International, Durham, NC, USA
| | - Eric Hillpot
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Lei Zhang
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Maria Zagorulya
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Greg Tombline
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Michael Gilbert
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samantha L Yuen
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Alexis Pope
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | | | - Stephan Emmrich
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Denis Firsanov
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Advait Athreya
- Department of Biology, University of Rochester, Rochester, NY, USA
| | | | - Jeehae Han
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Seungjin Ryu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Archana Tare
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yizhou Zhu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Adam Hudgins
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gil Atzmon
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Nir Barzilai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | - Benjamin A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics and Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA
| |
Collapse
|
31
|
Tripathi R, Gupta R, Sahu M, Srivastava D, Das A, Ambasta RK, Kumar P. Free radical biology in neurological manifestations: mechanisms to therapeutics interventions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62160-62207. [PMID: 34617231 DOI: 10.1007/s11356-021-16693-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Recent advancements and growing attention about free radicals (ROS) and redox signaling enable the scientific fraternity to consider their involvement in the pathophysiology of inflammatory diseases, metabolic disorders, and neurological defects. Free radicals increase the concentration of reactive oxygen and nitrogen species in the biological system through different endogenous sources and thus increased the overall oxidative stress. An increase in oxidative stress causes cell death through different signaling mechanisms such as mitochondrial impairment, cell-cycle arrest, DNA damage response, inflammation, negative regulation of protein, and lipid peroxidation. Thus, an appropriate balance between free radicals and antioxidants becomes crucial to maintain physiological function. Since the 1brain requires high oxygen for its functioning, it is highly vulnerable to free radical generation and enhanced ROS in the brain adversely affects axonal regeneration and synaptic plasticity, which results in neuronal cell death. In addition, increased ROS in the brain alters various signaling pathways such as apoptosis, autophagy, inflammation and microglial activation, DNA damage response, and cell-cycle arrest, leading to memory and learning defects. Mounting evidence suggests the potential involvement of micro-RNAs, circular-RNAs, natural and dietary compounds, synthetic inhibitors, and heat-shock proteins as therapeutic agents to combat neurological diseases. Herein, we explain the mechanism of free radical generation and its role in mitochondrial, protein, and lipid peroxidation biology. Further, we discuss the negative role of free radicals in synaptic plasticity and axonal regeneration through the modulation of various signaling molecules and also in the involvement of free radicals in various neurological diseases and their potential therapeutic approaches. The primary cause of free radical generation is drug overdosing, industrial air pollution, toxic heavy metals, ionizing radiation, smoking, alcohol, pesticides, and ultraviolet radiation. Excessive generation of free radicals inside the cell R1Q1 increases reactive oxygen and nitrogen species, which causes oxidative damage. An increase in oxidative damage alters different cellular pathways and processes such as mitochondrial impairment, DNA damage response, cell cycle arrest, and inflammatory response, leading to pathogenesis and progression of neurodegenerative disease other neurological defects.
Collapse
Affiliation(s)
- Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Ankita Das
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India.
- , Delhi, India.
- Molecular Neuroscience and Functional Genomics Laboratory, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
32
|
Cai J, Wang T, Zhou Y, Tang C, Liu Y, Dong Z. Phosphorylation by GSK-3β increases the stability of SIRT6 to alleviate TGF-β-induced fibrotic response in renal tubular cells. Life Sci 2022; 308:120914. [PMID: 36057401 DOI: 10.1016/j.lfs.2022.120914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 10/31/2022]
Abstract
AIMS The deacetylase Sirtuin 6 (SIRT6) is up-regulated during fibrogenesis in renal tubular cells and post-ischemia/reperfusion kidneys. Hence, our aim was to investigate the mechanism of SIRT6 up-regulation upon profibrotic stress. MAIN METHODS Immunohistochemical staining was used to detect the expression of UBC9 in the kidney section. The interaction of GSK-3β and SIRT6, and phosphorylation level of SIRT6 were detected by the immunoprecipitation assay. The wild-type and phosphorylated site mutant plasmids of SIRT6 were constructed and stably transfected to BUMPT cells to evaluate the phosphorylation function of SIRT6 by immunoblotting assay. KEY FINDINGS The phosphorylation of SIRT6 is significantly increased during TGF-β treatment in mouse renal tubular cells. GSK-3β can physically interact with SIRT6 in renal tubular cells, and this interaction is enhanced by TGF-β treatment. Moreover, GSK-3β is the phosphorylation kinase for SIRT6, and phosphorylates SIRT6 at Serine 326 residue to prevent its ubiquitination-mediated proteasomal degradation. Non-phosphorylatable mutant, S326A, of SIRT6, restores β-catenin activation and fibrotic changes in renal tubular cells. SIGNIFICANCE The present study demonstrates that a new mechanism for GSK-3β-mediated anti-fibrotic function in renal fibrosis through phosphorylation of SIRT6 to prevent its proteasomal degradation.
Collapse
Affiliation(s)
- Juan Cai
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Tianshi Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunqian Zhou
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China.
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
33
|
Wu Q, Gao ZJ, Yu X, Wang P. Dietary regulation in health and disease. Signal Transduct Target Ther 2022; 7:252. [PMID: 35871218 PMCID: PMC9308782 DOI: 10.1038/s41392-022-01104-w] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/21/2022] [Accepted: 07/04/2022] [Indexed: 02/08/2023] Open
Abstract
Nutriments have been deemed to impact all physiopathologic processes. Recent evidences in molecular medicine and clinical trials have demonstrated that adequate nutrition treatments are the golden criterion for extending healthspan and delaying ageing in various species such as yeast, drosophila, rodent, primate and human. It emerges to develop the precision-nutrition therapeutics to slow age-related biological processes and treat diverse diseases. However, the nutritive advantages frequently diversify among individuals as well as organs and tissues, which brings challenges in this field. In this review, we summarize the different forms of dietary interventions extensively prescribed for healthspan improvement and disease treatment in pre-clinical or clinical. We discuss the nutrient-mediated mechanisms including metabolic regulators, nutritive metabolism pathways, epigenetic mechanisms and circadian clocks. Comparably, we describe diet-responsive effectors by which dietary interventions influence the endocrinic, immunological, microbial and neural states responsible for improving health and preventing multiple diseases in humans. Furthermore, we expatiate diverse patterns of dietotheroapies, including different fasting, calorie-restricted diet, ketogenic diet, high-fibre diet, plants-based diet, protein restriction diet or diet with specific reduction in amino acids or microelements, potentially affecting the health and morbid states. Altogether, we emphasize the profound nutritional therapy, and highlight the crosstalk among explored mechanisms and critical factors to develop individualized therapeutic approaches and predictors.
Collapse
Affiliation(s)
- Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zhi-Jie Gao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Xin Yu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
34
|
Ruszkiewicz JA, Bürkle A, Mangerich A. Fueling genome maintenance: On the versatile roles of NAD + in preserving DNA integrity. J Biol Chem 2022; 298:102037. [PMID: 35595095 PMCID: PMC9194868 DOI: 10.1016/j.jbc.2022.102037] [Citation(s) in RCA: 9] [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: 01/19/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
NAD+ is a versatile biomolecule acting as a master regulator and substrate in various cellular processes, including redox regulation, metabolism, and various signaling pathways. In this article, we concisely and critically review the role of NAD+ in mechanisms promoting genome maintenance. Numerous NAD+-dependent reactions are involved in the preservation of genome stability, the cellular DNA damage response, and other pathways regulating nucleic acid metabolism, such as gene expression and cell proliferation pathways. Of note, NAD+ serves as a substrate to ADP-ribosyltransferases, sirtuins, and potentially also eukaryotic DNA ligases, all of which regulate various aspects of DNA integrity, damage repair, and gene expression. Finally, we critically analyze recent developments in the field as well as discuss challenges associated with therapeutic actions intended to raise NAD+ levels.
Collapse
Affiliation(s)
- Joanna A Ruszkiewicz
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany.
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany.
| | - Aswin Mangerich
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
35
|
Wu S, Liu H. Sirtuins-Novel Regulators of Epigenetic Alterations in Airway Inflammation. Front Genet 2022; 13:862577. [PMID: 35620467 PMCID: PMC9127257 DOI: 10.3389/fgene.2022.862577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Histone modification is an important epigenetic alteration, and histone deacetylases are involved in the occurrence and development of various respiratory diseases. Sirtuins (SIRTs) have been demonstrated to play an important role in the formation and progression of chronic inflammatory diseases of the respiratory tract. SIRTs participate in the regulation of oxidative stress and inflammation and are related to cell structure and cellular localization. This paper summarizes the roles and mechanisms of SIRTs in airway inflammation and describes the latest research on SIRT modulators, aiming to provide a theoretical basis for the study of potential epigenetic alteration-inducing drug targets.
Collapse
Affiliation(s)
- Shunyu Wu
- Department of Otolaryngological, the Second Affiliated Hospital of the Naval Military Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Huanhai Liu
- Department of Otolaryngological, the Second Affiliated Hospital of the Naval Military Medical University (Shanghai Changzheng Hospital), Shanghai, China
| |
Collapse
|
36
|
Yang Y, Zhu M, Liang J, Wang H, Sun D, Li H, Chen L. SIRT6 mediates multidimensional modulation to maintain organism homeostasis. J Cell Physiol 2022; 237:3205-3221. [PMID: 35621134 DOI: 10.1002/jcp.30791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 05/16/2022] [Indexed: 12/20/2022]
Abstract
As a member of the silent information regulators (sirtuins) family, SIRT6 can regulate a variety of biological processes, including DNA repair, glucose and lipid metabolism, oxidative stress and lifespan, and so forth. SIRT6 maintains organism homeostasis in a variety of phenotypes by mediating epigenetic regulation and posttranslational modification of functional proteins. In this review, we outline the structural basis of SIRT6 enzyme activity and its mechanism of maintaining organism homeostasis in a variety of phenotypes, with an emphasis on the upstream that regulates SIRT6 expression and the downstream substrates. And how SIRT6 achieves multidimensional coordination to maintain organism homeostasis and even extend lifespan. We try to understand the regulatory mechanism of SIRT6 in different phenotypes from the perspective of protein interaction.
Collapse
Affiliation(s)
- Yueying Yang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Man Zhu
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Jing Liang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Hui Wang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China.,School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
37
|
Zhou W, Xu Y, Zhang J, Zhang P, Yao Z, Yan Z, Wang H, Chu J, Yao S, Zhao S, Yang S, Guo Y, Miao J, Liu K, Chan WC, Xia Q, Liu Y. MiRNA-363-3p/DUSP10/JNK axis mediates chemoresistance by enhancing DNA damage repair in diffuse large B-cell lymphoma. Leukemia 2022; 36:1861-1869. [PMID: 35488020 PMCID: PMC9252898 DOI: 10.1038/s41375-022-01565-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022]
Abstract
Anthracycline-based chemotherapy resistance represents a major challenge in diffuse large B-cell lymphoma (DLBCL). MiRNA and gene expression profiles (n = 47) were determined to uncover potential chemoresistance mechanisms and therapeutic approaches. An independent correlation between high expression of miRNA-363-3p and chemoresistance was observed and validated in a larger cohort (n = 106). MiRNA-363-3p was shown to reduce doxorubicin-induced apoptosis and tumor shrinkage in in vitro and in vivo experiments by ectopic expression and CRISPR/Cas9-mediated knockout in DLBCL cell lines. DNA methylation was found to participate in transcriptional regulation of miRNA-363-3p. Further investigation revealed that dual specificity phosphatase 10 (DUSP10) is a target of miRNA-363-3p and its suppression promotes the phosphorylation of c-Jun N-terminal kinase (JNK). The miRNA-363-3p/DUSP10/JNK axis was predominantly associated with negative regulation of homologous recombination (HR) and DNA repair pathways. Ectopic expression of miRNA-363-3p more effectively repaired doxorubicin-induced double-strand break (DSB) while enhancing non-homologous end joining repair and reducing HR repair. Targeting JNK and poly (ADP-ribose) polymerase 1 significantly inhibited doxorubicin-induced DSB repair, increased doxorubicin-induced cell apoptosis and tumor shrinkage, and improved the survival of tumor-bearing mice. In conclusion, the miRNA-363-3p/DUSP10/JNK axis is a novel chemoresistance mechanism in DLBCL that may be reversed by targeted therapy.
Collapse
Affiliation(s)
- Wenping Zhou
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China.,Department of Lymphoma Research, Henan Cancer Institute, Zhengzhou, Henan, China
| | - Yuanlin Xu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jiuyang Zhang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Peipei Zhang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China.,Department of Lymphoma Research, Henan Cancer Institute, Zhengzhou, Henan, China
| | - Zhihua Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zheng Yan
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Haiying Wang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Junfeng Chu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Shuna Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Shuang Zhao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Shujun Yang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yongjun Guo
- Department of Molecule and Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Wing C Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Qingxin Xia
- Department of Molecule and Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China.
| | - Yanyan Liu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China. .,Department of Lymphoma Research, Henan Cancer Institute, Zhengzhou, Henan, China.
| |
Collapse
|
38
|
Li Y, Jin J, Wang Y. SIRT6 Widely Regulates Aging, Immunity, and Cancer. Front Oncol 2022; 12:861334. [PMID: 35463332 PMCID: PMC9019339 DOI: 10.3389/fonc.2022.861334] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/14/2022] [Indexed: 12/14/2022] Open
Abstract
SIRT6 is a member of the Sir2-like family in mammals. Recent structural and biochemical studies have characterized SIRT6 as having deacetylation, defatty-acylation, and mono-ADP-ribosylation activities, which determine its important regulatory roles during physiological and pathological processes. This review focuses mainly on the regulatory functions of SIRT6 in aging, cancer, and, especially, immunity. Particular attention is paid to studies illustrating the critical role of SIRT6 in the regulation of immune cells from the viewpoints of immunesenescence, immunometabolism, and tumor immunology. Owing to its role in regulating the function of the immune system, SIRT6 can be considered to be a potential therapeutic target for the treatment of diseases.
Collapse
Affiliation(s)
- Yunjia Li
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Heifei, China
| | - Jing Jin
- Institute of Immunology and the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, China
| | - Yi Wang
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Heifei, China.,Institute of Immunology and the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, China
| |
Collapse
|
39
|
Natural Bioactive Compounds Targeting Histone Deacetylases in Human Cancers: Recent Updates. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082568. [PMID: 35458763 PMCID: PMC9027183 DOI: 10.3390/molecules27082568] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/13/2022]
Abstract
Cancer is a complex pathology that causes a large number of deaths worldwide. Several risk factors are involved in tumor transformation, including epigenetic factors. These factors are a set of changes that do not affect the DNA sequence, while modifying the gene’s expression. Histone modification is an essential mark in maintaining cellular memory and, therefore, loss of this mark can lead to tumor transformation. As these epigenetic changes are reversible, the use of molecules that can restore the functions of the enzymes responsible for the changes is therapeutically necessary. Natural molecules, mainly those isolated from medicinal plants, have demonstrated significant inhibitory properties against enzymes related to histone modifications, particularly histone deacetylases (HDACs). Flavonoids, terpenoids, phenolic acids, and alkaloids exert significant inhibitory effects against HDAC and exhibit promising epi-drug properties. This suggests that epi-drugs against HDAC could prevent and treat various human cancers. Accordingly, the present study aimed to evaluate the pharmacodynamic action of different natural compounds extracted from medicinal plants against the enzymatic activity of HDAC.
Collapse
|
40
|
SIRT6 mediates MRTF-A deacetylation in vascular endothelial cells to antagonize oxLDL-induced ICAM-1 transcription. Cell Death Dis 2022; 8:96. [PMID: 35246513 PMCID: PMC8897425 DOI: 10.1038/s41420-022-00903-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/10/2022] [Accepted: 02/03/2022] [Indexed: 01/01/2023]
Abstract
Oxidized low-density lipoprotein (oxLDL), a known risk factor for atherosclerosis, activates the transcription of adhesion molecules (ICAM-1) in endothelial cells. We previously showed that myocardin-related transcription factor A (MRTF-A) mediates oxLDL-induced ICAM-1 transcription. Here we confirm that ICAM-1 transactivation paralleled dynamic alterations in MRTF-A acetylation. Since treatment with the antioxidant NAC dampened MRTF-A acetylation, MRTF-A acetylation appeared to be sensitive to cellular redox status. Of interest, silencing of SIRT6, a lysine deacetylase, restored MRTF-A acetylation despite the addition of NAC. SIRT6 directly interacted with MRTF-A to modulate MRTF-A acetylation. Deacetylation of MRTF-A by SIRT6 led to its nuclear expulsion thus dampening MRTF-A occupancy on the ICAM-1 promoter. Moreover, SIRT6 expression was downregulated with oxLDL stimulation likely owing to promoter hypermethylation in endothelial cells. DNA methyltransferase 1 (DNMT1) was recruited to the SIRT6 promoter and mediated SIRT6 repression. The ability of DNMT1 to repress SIRT6 promoter partly was dependent on ROS-sensitive serine 154 phosphorylation. In conclusion, our data unveil a novel DNMT1-SIRT6 axis that contributes to the regulation of MRTF-A acetylation and ICAM-1 transactivation in endothelial cells.
Collapse
|
41
|
Modulation of SIRT6 activity acts as an emerging therapeutic implication for pathological disorders in the skeletal system. Genes Dis 2022. [DOI: 10.1016/j.gendis.2021.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
42
|
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol 2022; 23:56-73. [PMID: 34518687 PMCID: PMC8692439 DOI: 10.1038/s41580-021-00411-4] [Citation(s) in RCA: 361] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 02/08/2023]
Abstract
Dietary restriction with adequate nutrition is the gold standard for delaying ageing and extending healthspan and lifespan in diverse species, including rodents and non-human primates. In this Review, we discuss the effects of dietary restriction in these mammalian model organisms and discuss accumulating data that suggest that dietary restriction results in many of the same physiological, metabolic and molecular changes responsible for the prevention of multiple ageing-associated diseases in humans. We further discuss how different forms of fasting, protein restriction and specific reductions in the levels of essential amino acids such as methionine and the branched-chain amino acids selectively impact the activity of AKT, FOXO, mTOR, nicotinamide adenine dinucleotide (NAD+), AMP-activated protein kinase (AMPK) and fibroblast growth factor 21 (FGF21), which are key components of some of the most important nutrient-sensing geroprotective signalling pathways that promote healthy longevity.
Collapse
Affiliation(s)
- Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
- Department of Clinical and Experimental Sciences, Brescia University School of Medicine, Brescia, Italy.
| |
Collapse
|
43
|
Xu H, Liu YY, Li LS, Liu YS. Sirtuins at the Crossroads between Mitochondrial Quality Control and Neurodegenerative Diseases: Structure, Regulation, Modifications, and Modulators. Aging Dis 2022; 14:794-824. [PMID: 37191431 DOI: 10.14336/ad.2022.1123] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/23/2022] [Indexed: 04/03/2023] Open
Abstract
Sirtuins (SIRT1-SIRT7), a family of nicotinamide adenine dinucleotide (NAD+)-dependent enzymes, are key regulators of life span and metabolism. In addition to acting as deacetylates, some sirtuins have the properties of deacylase, decrotonylase, adenosine diphosphate (ADP)-ribosyltransferase, lipoamidase, desuccinylase, demalonylase, deglutarylase, and demyristolyase. Mitochondrial dysfunction occurs early on and acts causally in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Sirtuins are implicated in the regulation of mitochondrial quality control, which is highly associated with the pathogenesis of neurodegenerative diseases. There is growing evidence indicating that sirtuins are promising and well-documented molecular targets for the treatment of mitochondrial dysfunction and neurodegenerative disorders by regulating mitochondrial quality control, including mitochondrial biogenesis, mitophagy, mitochondrial fission/fusion dynamics, and mitochondrial unfolded protein responses (mtUPR). Therefore, elucidation of the molecular etiology of sirtuin-mediated mitochondrial quality control points to new prospects for the treatment of neurodegenerative diseases. However, the mechanisms underlying sirtuin-mediated mitochondrial quality control remain obscure. In this review, we update and summarize the current understanding of the structure, function, and regulation of sirtuins with an emphasis on the cumulative and putative effects of sirtuins on mitochondrial biology and neurodegenerative diseases, particularly their roles in mitochondrial quality control. In addition, we outline the potential therapeutic applications for neurodegenerative diseases of targeting sirtuin-mediated mitochondrial quality control through exercise training, calorie restriction, and sirtuin modulators in neurodegenerative diseases.
Collapse
|
44
|
Raghu S, Prabhashankar AB, Shivanaiah B, Tripathi E, Sundaresan NR. Sirtuin 6 Is a Critical Epigenetic Regulator of Cancer. Subcell Biochem 2022; 100:337-360. [PMID: 36301499 DOI: 10.1007/978-3-031-07634-3_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Sirtuin 6 (SIRT6) is a member of the mammalian sirtuin family with deacetylase, deacylase, and mono-ADP-ribosyl-transferase activities. It is a multitasking chromatin-associated protein regulating different cellular and physiological functions in cells. Specifically, SIRT6 dysfunction is implicated in several aging-related human diseases, including cancer. Studies indicate that SIRT6 has a tumor-specific role, and it is considered a tumor suppressor as well as a tumor growth inducer, depending on the type of cancer. In this chapter, we review the role of SIRT6 in metabolism, genomic stability, and cancer. Further, we provide an insight into the interplay of the tumor-suppressing and oncogenic roles of SIRT6 in cancer. Additionally, we discuss the use of small-molecule SIRT6 modulators as potential therapeutics.
Collapse
Affiliation(s)
- Sukanya Raghu
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India
| | - Arathi Bangalore Prabhashankar
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India
| | - Bhoomika Shivanaiah
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India
| | - Ekta Tripathi
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, Ramaiah University of Applied Sciences, Bengaluru, India.
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India.
| | - Nagalingam Ravi Sundaresan
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science (IISc), Bengaluru, Karnataka, India.
| |
Collapse
|
45
|
Yang X, Feng J, Liang W, Zhu Z, Chen Z, Hu J, Yang D, Ding G. Roles of SIRT6 in kidney disease: a novel therapeutic target. Cell Mol Life Sci 2021; 79:53. [PMID: 34950960 PMCID: PMC11072764 DOI: 10.1007/s00018-021-04061-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022]
Abstract
SIRT6 is an NAD+ dependent deacetylase that belongs to the mammalian sirtuin family. SIRT6 is mainly located in the nucleus and regulates chromatin remodeling, genome stability, and gene transcription. SIRT6 extensively participates in various physiological activities such as DNA repair, energy metabolism, oxidative stress, inflammation, and fibrosis. In recent years, the role of epigenetics such as acetylation modification in renal disease has gradually received widespread attention. SIRT6 reduces oxidative stress, inflammation, and renal fibrosis, which is of great importance in maintaining cellular homeostasis and delaying the chronic progression of kidney disease. Here, we review the structure and biological function of SIRT6 and summarize the regulatory mechanisms of SIRT6 in kidney disease. Moreover, the role of SIRT6 as a potential therapeutic target for the progression of kidney disease will be discussed. SIRT6 plays an important role in kidney disease. SIRT6 regulates mitochondrial dynamics and mitochondrial biogenesis, induces G2/M cycle arrest, and plays an antioxidant role in nephrotoxicity, IR, obstructive nephropathy, and sepsis-induced AKI. SIRT6 prevents and delays progressive CKD induced by hyperglycemia, kidney senescence, hypertension, and lipid accumulation by regulating mitochondrial biogenesis, and has antioxidant, anti-inflammatory, and antifibrosis effects. Additionally, hypoxia, inflammation, and fibrosis are the main mechanisms of the AKI-to-CKD transition. SIRT6 plays a critical role in the AKI-to-CKD transition and kidney repair through anti-inflammatory, antifibrotic, and mitochondrial quality control mechanisms. AKI Acute kidney injury, CKD Chronic kidney disease.
Collapse
Affiliation(s)
- Xueyan Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Dingping Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
46
|
Korotkov A, Seluanov A, Gorbunova V. Sirtuin 6: linking longevity with genome and epigenome stability. Trends Cell Biol 2021; 31:994-1006. [PMID: 34281779 PMCID: PMC8903056 DOI: 10.1016/j.tcb.2021.06.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 01/13/2023]
Abstract
Sirtuin 6 (SIRT6) has been in the spotlight of aging research because progeroid phenotypes are associated with SIRT6 deficiency. SIRT6 has multiple molecular functions, including DNA repair and heterochromatin regulation, which position SIRT6 as a hub that regulates genome and epigenome stability. Genomic instability caused by persistent DNA damage and accumulating mutations, together with alterations in the epigenetic landscape and derepression of repetitive genetic elements, have emerged as mechanisms driving organismal aging. Enhanced levels of SIRT6 expression or activity provide avenues for rejuvenation strategies. This review focuses on the role of SIRT6 in the maintenance of genome and epigenome stability and its link to longevity. We propose a model where SIRT6 together with lamins control aging and rejuvenation by maintaining epigenetic silencing of repetitive elements.
Collapse
Affiliation(s)
- Anatoly Korotkov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA.
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA.
| |
Collapse
|
47
|
Li J, M. Saville K, Ibrahim M, Zeng X, McClellan S, Angajala A, Beiser A, Andrews JF, Sun M, Koczor CA, Clark J, Hayat F, Makarov MV, Wilk A, Yates NA, Migaud ME, Sobol RW. NAD + bioavailability mediates PARG inhibition-induced replication arrest, intra S-phase checkpoint and apoptosis in glioma stem cells. NAR Cancer 2021; 3:zcab044. [PMID: 34806016 PMCID: PMC8600031 DOI: 10.1093/narcan/zcab044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/20/2021] [Accepted: 11/10/2021] [Indexed: 01/31/2023] Open
Abstract
Elevated expression of the DNA damage response proteins PARP1 and poly(ADP-ribose) glycohydrolase (PARG) in glioma stem cells (GSCs) suggests that glioma may be a unique target for PARG inhibitors (PARGi). While PARGi-induced cell death is achieved when combined with ionizing radiation, as a single agent PARG inhibitors appear to be mostly cytostatic. Supplementation with the NAD+ precursor dihydronicotinamide riboside (NRH) rapidly increased NAD+ levels in GSCs and glioma cells, inducing PARP1 activation and mild suppression of replication fork progression. Administration of NRH+PARGi triggers hyperaccumulation of poly(ADP-ribose) (PAR), intra S-phase arrest and apoptosis in GSCs but minimal PAR induction or cytotoxicity in normal astrocytes. PAR accumulation is regulated by select PARP1- and PAR-interacting proteins. The involvement of XRCC1 highlights the base excision repair pathway in responding to replication stress while enhanced interaction of PARP1 with PCNA, RPA and ORC2 upon PAR accumulation implicates replication associated PARP1 activation and assembly with pre-replication complex proteins upon initiation of replication arrest, the intra S-phase checkpoint and the onset of apoptosis.
Collapse
Affiliation(s)
- Jianfeng Li
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Kate M. Saville
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Md Ibrahim
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Xuemei Zeng
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15213, USA
| | - Steve McClellan
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Anusha Angajala
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Alison Beiser
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Joel F Andrews
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Mai Sun
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15213, USA
| | - Christopher A Koczor
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Jennifer Clark
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Faisal Hayat
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Mikhail V Makarov
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Anna Wilk
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Nathan A Yates
- Biomedical Mass Spectrometry Center, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, PA 15213, USA,Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Marie E Migaud
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA,Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Robert W Sobol
- To whom correspondence should be addressed. Tel: +1 251 445 9846;
| |
Collapse
|
48
|
Gupta R, Ambasta RK, Kumar P. Multifaced role of protein deacetylase sirtuins in neurodegenerative disease. Neurosci Biobehav Rev 2021; 132:976-997. [PMID: 34742724 DOI: 10.1016/j.neubiorev.2021.10.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 01/07/2023]
Abstract
Sirtuins, a class III histone/protein deacetylase, is a central regulator of metabolic function and cellular stress response. This plays a pivotal role in the pathogenesis and progression of diseases such as cancer, neurodegeneration, metabolic syndromes, and cardiovascular disease. Sirtuins regulate biological and cellular processes, for instance, mitochondrial biogenesis, lipid and fatty acid oxidation, oxidative stress, gene transcriptional activity, apoptosis, inflammatory response, DNA repair mechanism, and autophagic cell degradation, which are known components for the progression of the neurodegenerative diseases (NDDs). Emerging evidence suggests that sirtuins are the useful molecular targets against NDDs like, Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD), and Amyotrophic Lateral Sclerosis (ALS). However, the exact mechanism of neuroprotection mediated through sirtuins remains unsettled. The manipulation of sirtuins activity with its modulators, calorie restriction (CR), and micro RNAs (miR) is a novel therapeutic approach for the treatment of NDDs. Herein, we reviewed the current putative therapeutic role of sirtuins in regulating synaptic plasticity and cognitive functions, which are mediated through the different molecular phenomenon to prevent neurodegeneration. We also explained the implications of sirtuin modulators, and miR based therapies for the treatment of life-threatening NDDs.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
49
|
Brito VB, Nascimento LVM, Moura DJ, Saffi J. Cardioprotective Effect of Maternal Supplementation with Resveratrol on Toxicity Induced by Doxorubicin in Offspring Cardiomyocytes. Arq Bras Cardiol 2021; 117:1147-1158. [PMID: 34644787 PMCID: PMC8757151 DOI: 10.36660/abc.20200752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/27/2021] [Indexed: 11/28/2022] Open
Abstract
Fundamento A doxorrubicina (DOX) é frequentemente usada para tratar muitos tipos de cânceres, apesar da cardiotoxicidade dose-dependente. Como alternativa, o resveratrol é um polifenol que tem demonstrado efeitos cardioprotetores em vários modelos de disfunção cardíaca. Objetivo Este estudo investigou se o tratamento com resveratrol em ratas gestantes protege contra toxicidade induzida por doxorrubicina em cardiomiócitos da ninhada. Métodos Ratas Wistar (n-8) receberam sresveratrol como suplemento alimentar durante a gestação. No nascimento da ninhada, os corações (9-11) foram usados para se obter a cultura primária de cardiomiócitos. A cardiotoxicidade induzida por DOX e os efeitos da suplementação com resveratrol foram avaliados por marcadores de stress oxidativo, tais como oxidação da diclorofluoresceína diacetato, diminuição da atividade de enzimas antioxidantes, e oxidação do teor total de grupos sulfidrila, além da avaliação da viabilidade celular, geração de danos ao DNA, bem como a resposta de reparo aos danos ao DNA. Um valor de p <0,05 foi considerado estatisticamente significativo. Resultados Os cardiomiócitos de neonatos de ratas que receberam suplemento resveratrol apresentaram um aumento (p <0,01) na viabilidade das células, e diminuição (p <0,0001) de células apoptóticas/necróticas após o tratamento com DOX, o que está correlacionado às atividades de enzimas antioxidantes e produção de diclorofluoresceína. Além disso, o resveratrol protegeu os cardiomiócitos de danos ao DNA induzidos por DOX, apresentando uma diminuição (p <0,05) nas quebras de DNA induzidas por stress oxidativo, avaliadas pela atividade de enzimas reparadoras do DNA endonuclease III e formamidopirimidina glicosilase. A suplementação com resveratrol aumentou (p <0,05) a expressão da proteína reparadora Sirt6 nos cardiomiócitos dos filhotes. Conclusão Essa pesquisa indica que a suplementação com resveratrol durante o período gestacional tem um efeito cardioprotetor no coração da ninhada contra a toxicidade induzida por DOX, o que pode se dever a sua função antioxidante, e o aumento na resposta de danos ao DNA.
Collapse
Affiliation(s)
- Verônica Bidinotto Brito
- Universidade Federal de Ciências da Saúde de Porto Alegre , Porto Alegre , RS - Brasil.,Faculdades Integradas de Taquara , Taquara , RS - Brasil
| | | | | | - Jenifer Saffi
- Universidade Federal de Ciências da Saúde de Porto Alegre , Porto Alegre , RS - Brasil
| |
Collapse
|
50
|
Hai R, He L, Shu G, Yin G. Characterization of Histone Deacetylase Mechanisms in Cancer Development. Front Oncol 2021; 11:700947. [PMID: 34395273 PMCID: PMC8360675 DOI: 10.3389/fonc.2021.700947] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/05/2021] [Indexed: 02/01/2023] Open
Abstract
Over decades of studies, accumulating evidence has suggested that epigenetic dysregulation is a hallmark of tumours. Post-translational modifications of histones are involved in tumour pathogenesis and development mainly by influencing a broad range of physiological processes. Histone deacetylases (HDACs) and histone acetyltransferases (HATs) are pivotal epigenetic modulators that regulate dynamic processes in the acetylation of histones at lysine residues, thereby influencing transcription of oncogenes and tumour suppressor genes. Moreover, HDACs mediate the deacetylation process of many nonhistone proteins and thus orchestrate a host of pathological processes, such as tumour pathogenesis. In this review, we elucidate the functions of HDACs in cancer.
Collapse
Affiliation(s)
- Rihan Hai
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.,School of Basic Medical Sciences, Central South University, Changsha, China
| | - Liuer He
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China.,School of Basic Medical Sciences, Central South University, Changsha, China
| | - Guang Shu
- School of Basic Medical Sciences, Central South University, Changsha, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|