1
|
Hao Y, Hu J, Zhang Z, Guan Q, Wang J, Tao Y, Cheng J, Fan Y. Sirt6 deficiency exacerbates angiotensin II-induced lipid nephrotoxicity by affecting PLD6-derived cardiolipin metabolism in podocytes. Cell Signal 2025; 133:111858. [PMID: 40355014 DOI: 10.1016/j.cellsig.2025.111858] [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: 12/21/2024] [Revised: 04/22/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND AIMS Perturbation of cardiolipin (CL) metabolism is associated with lipid nephrotoxicity. Recent findings provide new insights into the roles of CL-modulating proteins as critical determinants of podocyte function in chronic kidney disease (CKD). We previously demonstrated that Sirtuin 6 (Sirt6) is a compelling target inhibiting Angiotensin II (Ang II)-induced lipid dysregulation in podocytes. However, whether Sirt6 regulates podocyte CL metabolism is unknown. METHODS Renal biopsy specimens of patients with hypertensive nephropathy (HN) were used in this study. Podocyte Sirt6-specific knockout mice were generated using the Cre-loxP system. The effect of Sirt6 on mitochondrial CL metabolism, especially the peroxidation and hydrolysis of CL, was investigated in Ang II-infusion mice and Ang II-induced cultured podocytes. RESULTS Sirt6 and outer mitochondrial membrane protein phospholipase D family member 6 (PLD6) were decreased in the glomeruli of patients with HN. Ang II downregulated Sirt6 and PLD6 expression in podocytes in vitro and in vivo. Podocyte-specific deletion of Sirt6 exacerbated lipid droplets formation, CL accumulation and peroxidation, aggravated Ang II-induced mitochondrial dysfunction and cell apoptosis. Mechanically, Sirt6 maintained podocyte CL homeostasis, at least in part through PLD6 signaling-mediated CL metabolism. In addition, cardiolipin antioxidant Szeto-Schiller Peptide 31 (SS-31) treatment inhibited Ang II-induced lipid accumulation and CL peroxidation in podocytes. CONCLUSIONS Our findings shed light on Sirt6's regulatory mechanisms on podocyte CL metabolism and suggest exploiting the Sirt6-PLD6 axis as a potential therapeutic target for protecting against lipid nephrotoxicity.
Collapse
Affiliation(s)
- Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Qian Guan
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Juan Wang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yu Tao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Yanqin Fan
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Wang J, Ren Y, Qu S. Modulation of Sirtuins to address aging related disorders through the use of selected phytochemicals. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156648. [PMID: 40359853 DOI: 10.1016/j.phymed.2025.156648] [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: 12/26/2024] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Aging is a complex phenomenon involving oxidative stress, inflammation, and cellular damage subsequently leading to various disorders, such as cardiovascular diseases, neurodegenerative disorders, diabetes, and cancer. Sirtuin (SIRT) proteins are one of the major molecular factors that affect human aging. Sirtuins are a class of NAD+-dependent enzymes that control oxidative stress response, DNA damage repair, inflammation and metabolism, all of which are involved in aging and age-related diseases. PURPOSE The objective of this review is to elucidate the potential role of SIRT in the aging process and modulation of SIRT pathway through selected phytochemicals like Curcumin, Resveratrol, Quercetin, and Kaempferol. RESULTS Studies convincedly revealed that SIRT pathway represents a promising avenue for extending the human health span and addressing age-related conditions. Phytochemicals like Curcumin, Resveratrol, Quercetin, and Kaempferol have shown excellent potential to mediate aging effects through their potent antioxidant, anti-inflammatory, and regulatory activities. These potent bioactive compounds enhance oxidative stress response, genomic integrity, neuroprotective and anti-inflammatory activities through SIRT pathway modulation. Furthermore, in addition to antiaging effects, other therapeutic benefits are also associated with each compound including nervous disorders, cancer, and metabolic disorders are also briefly highlighted. Studies reported convincing evidence that Curcumin, Resveratrol, Quercetin, and Kaempferol, effectively modulate SIRT expression/activity leading to improved cell stress tolerance, reduced oxidation and enhanced metabolic state. CONCLUSIONS Collectively, studies revealed the comprehensive nutraceutical significance of Curcumin, Resveratrol, Quercetin, and Kaempferol as anti-aging therapeutics and warrant future studies to exploit the full potential of these natural compounds.
Collapse
Affiliation(s)
- Jing Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University. 36 Sanhao Street, Heping District, 110004 Shenyang, Liaoning, PR China.
| | - Yaoyao Ren
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004 Shenyang, Liaoning, PR China.
| | - Shengtao Qu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, No. 36 Sanhao St, Shenyang 110004, PR China.
| |
Collapse
|
3
|
Wen T, Chen W, Wang F, Zhang R, Chen C, Zhang M, Ma T. The roles and functions of ergothioneine in metabolic diseases. J Nutr Biochem 2025; 141:109895. [PMID: 40058711 DOI: 10.1016/j.jnutbio.2025.109895] [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: 10/18/2024] [Revised: 01/25/2025] [Accepted: 03/04/2025] [Indexed: 04/04/2025]
Abstract
The global prevalence of metabolic diseases is on the increase, and it has become a significant threat to the health and lives of individuals. Ergothioneine (EGT) is a natural betaine amino acid found in various foods, particularly mushrooms. EGT cannot be synthesized by mammals; it is absorbed into small intestinal epithelial cells by a cationic protein, the novel organic cation transporter 1 (OCTN1), and transported to certain organs including liver, spleen, kidney, lung, heart, eyes and brain. EGT has been reported to exhibit antioxidant, anti-inflammatory, anti-apoptotic, anti-aging, and metal-chelating effects. The unique chemical properties and biological functions of EGT position it as a promising candidate for the research and treatment of metabolic diseases. This review summarizes EGT's capacities, potential therapeutic effects on multiple metabolic diseases, and their specific mechanisms. Finally, we outline challenges for future research on EGT and aspire to establish it as a prospective therapeutic agent for metabolic diseases.
Collapse
Affiliation(s)
- Tingting Wen
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Wanjing Chen
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Fengjing Wang
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Rui Zhang
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Cheng Chen
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China.
| | - Mingliang Zhang
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Teng Ma
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
4
|
Greer EL, Lee SS, Prahlad V. Chromatin and epigenetics in aging biology. Genetics 2025; 230:iyaf055. [PMID: 40202900 DOI: 10.1093/genetics/iyaf055] [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: 06/20/2024] [Accepted: 02/03/2025] [Indexed: 04/11/2025] Open
Abstract
This book chapter will focus on modifications to chromatin itself, how chromatin modifications are regulated, and how these modifications are deciphered by the cell to impact aging. In this chapter, we will review how chromatin modifications change with age, examine how chromatin-modifying enzymes have been shown to regulate aging and healthspan, discuss how some of these epigenetic changes are triggered and how they can regulate the lifespan of the individual and its naïve descendants, and speculate on future directions for the field.
Collapse
Affiliation(s)
- Eric Lieberman Greer
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Siu Sylvia Lee
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Veena Prahlad
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
5
|
Yu F, Chen C, Liu W, Zhao Z, Fan Y, Li Z, Huang W, Xie T, Luo C, Yao Z, Guo Q, Yang Z, Liu J, Zhang Y, Kellems RE, Xia J, Li J, Xia Y. Longevity Humans Have Youthful Erythrocyte Function and Metabolic Signatures. Aging Cell 2025; 24:e14482. [PMID: 39924931 PMCID: PMC12074018 DOI: 10.1111/acel.14482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/30/2024] [Accepted: 12/21/2024] [Indexed: 02/11/2025] Open
Abstract
Longevity individuals have lower susceptibility to chronic hypoxia, inflammation, oxidative stress, and aging-related diseases. It has long been speculated that "rejuvenation molecules" exist in their blood to promote extended lifespan. We unexpectedly discovered that longevity individuals exhibit erythrocyte oxygen release function similar to young individuals, whereas most elderly show reduced oxygen release capacity. Untargeted erythrocyte metabolomics profiling revealed that longevity individuals are characterized by youth-like metabolic reprogramming and these metabolites effectively differentiate the longevity from the elderly. Quantification analyses led us to identify multiple novel longevity-related metabolites within erythrocytes including adenosine, sphingosine-1-phosphate (S1P), and glutathione (GSH) related amino acids. Mechanistically, we revealed that increased bisphosphoglycerate mutase (BPGM) and reduced MFSD2B protein levels in the erythrocytes of longevity individuals collaboratively work together to induce elevation of intracellular S1P, promote the release of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) from membrane to the cytosol, and thereby orchestrate glucose metabolic reprogramming toward Rapoport-Luebering Shunt to induce the 2,3-BPG production and trigger oxygen delivery. Furthermore, increased glutamine and glutamate transporter expression coupled with the enhanced intracellular metabolism underlie the elevated GSH production and the higher anti-oxidative stress capacity in the erythrocytes of longevity individuals. As such, longevity individuals displayed less systemic hypoxia-related metabolites and more antioxidative and anti-inflammatory metabolites in the plasma, thereby healthier clinical outcomes including lower inflammation parameters as well as better glucose-lipid metabolism, and liver and kidney function. Overall, we identified that youthful erythrocyte function and metabolism enable longevity individuals to better counteract peripheral tissue hypoxia, inflammation, and oxidative stress, thus maintaining healthspan.
Collapse
|
6
|
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] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [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
|
7
|
Wang X, Wang L, Zhou L, Chen L, Shi J, Ge J, Tian S, Yang Z, Zhou Y, Yu Q, Jin J, Ding C, Pan Y, Zou D. NUP62 alleviates senescence and promotes the stemness of human dental pulp stem cells via NSD2-dependent epigenetic reprogramming. Int J Oral Sci 2025; 17:34. [PMID: 40246825 PMCID: PMC12006529 DOI: 10.1038/s41368-025-00362-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Stem cells play a crucial role in maintaining tissue regenerative capacity and homeostasis. However, mechanisms associated with stem cell senescence require further investigation. In this study, we conducted a proteomic analysis of human dental pulp stem cells (HDPSCs) obtained from individuals of various ages. Our findings showed that the expression of NUP62 was decreased in aged HDPSCs. We discovered that NUP62 alleviated senescence-associated phenotypes and enhanced differentiation potential both in vitro and in vivo. Conversely, the knocking down of NUP62 expression aggravated the senescence-associated phenotypes and impaired the proliferation and migration capacity of HDPSCs. Through RNA-sequence and decoding the epigenomic landscapes remodeled induced by NUP62 overexpression, we found that NUP62 helps alleviate senescence in HDPSCs by enhancing the nuclear transport of the transcription factor E2F1. This, in turn, stimulates the transcription of the epigenetic enzyme NSD2. Finally, the overexpression of NUP62 influences the H3K36me2 and H3K36me3 modifications of anti-aging genes (HMGA1, HMGA2, and SIRT6). Our results demonstrated that NUP62 regulates the fate of HDPSCs via NSD2-dependent epigenetic reprogramming.
Collapse
Affiliation(s)
- Xiping Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Li Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Linxi Zhou
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lu Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiayi Shi
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jing Ge
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Sha Tian
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zihan Yang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yuqiong Zhou
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Qihao Yu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jiacheng Jin
- Touro College of Dental Medicine, New York Medical College, New York, USA
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yihuai Pan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
- Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Duohong Zou
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
- Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
8
|
Karmakar A, Augustine ABHR, Thummer RP. Genes as Genome Stabilizers in Pluripotent Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40095244 DOI: 10.1007/5584_2025_853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Pluripotent stem cells, comprising embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are characterized by their self-renewal capacity and the ability to differentiate into cells of all three germ layers of an adult animal. Out of the two, iPSCs are generated through the reprogramming of somatic cells by inducing a pluripotency-specific transcriptional program. This process requires a resetting of the somatic cell genome to a pluripotent cell-specific genome, resulting in cellular stress at genomic, epigenetic, and transcriptional levels. Notably, in contrast to the predominant compact and inactive organization of chromatin in somatic cells, the chromatin in ESCs and iPSCs is open. Furthermore, maintaining a pluripotent state needs a plethora of changes in the genetic landscape of the cells. Here, we attempt to elucidate how certain genes safeguard genomic stability in ESCs and iPSCs, aiding in the complex cellular mechanisms that regulate self-renewal, pluripotency, and somatic reprogramming.
Collapse
Affiliation(s)
- Asmita Karmakar
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Allan Blessing Harison Raj Augustine
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
9
|
Lv P, Hu Y, Ding L, Xiang W. Puerarin triggers sensitivity to ferroptosis in glioblastoma cells by activating SIRT3/NCOA4-dependent autophagy. Int Immunopharmacol 2025; 149:114246. [PMID: 39929095 DOI: 10.1016/j.intimp.2025.114246] [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: 09/09/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
SIRT3 has been found to involve in the tumorigenesis and progression of glioblastoma, and it is reported that puerarin can inhibit the growth of glioblastoma cells. Therefore, we aimed to investigate the biological function of SIRT3 in autophagy and ferroptosis in glioblastoma cells and study the effects of puerarin on ferroptosis and SIRT3/NCOA4-dependent autophagy in cancer cells. The results showed that overexpression of SIRT3 significantly promoted ferroptosis sensitization by reducing cell viability and GSH/GSSH ratio and increasing ROS levels, whereas knockout of SIRT3 significantly triggered cell viability and GSH/GSSH ratio and decreasing ROS levels in U87MG cells (P < 0.05). Moreover, overexpression of SIRT3 significantly also promoted the ratio of LC3-Ⅱ/Ⅰ and upregulated NCOA4 and Fe2+ levels, but downregulated the expression of p62 and FTH (P < 0.05); while knockout of SIRT3 has the opposite effects. Besides, autophagy inhibitor antagonized the effects of SIRT3 overexpression on the expression of autophagy-associated proteins and Fe2+ levels. Additionally, knockout of NCOA4 significantly increased cell viability and GSH/GSSH ratio and reduced ROS levels in RSL3-treated cells overexpressing SIRT3. Puerarin treatment significantly upregulated the levels of ROS, Fe2+, SIRT3, NCOA4, and the ratio of LC3-Ⅱ/Ⅰ, but downregulated the levels of cell viability, GSH/GSSH ratio, p62, and FTH in U87MG cells (all P < 0.05). Autophagy inhibitor, SIRT3 or NCOA4 deletion significantly reduced the effects of puerarin on the autophagy-dependent ferroptosis in U87MG cells (all P < 0.05). SIRT3 drives sensitivity to ferroptosis by activating NCOA4-mediated autophagy, and we proposed puerarin, a promising therapeutic drug for glioblastoma.
Collapse
Affiliation(s)
- Peng Lv
- Department of Neurosurgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China; Department of Neurosurgery Suizhou Central Hospital Hubei University of Medicine Suizhou Hubei China
| | - Yueyun Hu
- Department of Neurosurgery Wuhan No 8 Hospital Wuhan China
| | - Lei Ding
- General Hospital of the Yangtze River Shipping Wuhan Brain Hospital Wuhan China.
| | - Wei Xiang
- Department of Neurosurgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.
| |
Collapse
|
10
|
Gorbunova V, Seluanov A. SIRT5 slows skeletal muscle ageing by alleviating inflammation. Nat Metab 2025; 7:447-449. [PMID: 40087406 DOI: 10.1038/s42255-025-01228-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Affiliation(s)
- Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA.
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA.
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
11
|
Sun Y, Wang C, Wen L, Ling Z, Xia J, Cheng B, Peng J. Quercetin ameliorates senescence and promotes osteogenesis of BMSCs by suppressing the repetitive element‑triggered RNA sensing pathway. Int J Mol Med 2025; 55:4. [PMID: 39450556 PMCID: PMC11537266 DOI: 10.3892/ijmm.2024.5445] [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: 07/09/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Cell senescence impedes the self‑renewal and osteogenic capacity of bone marrow mesenchymal stem cells (BMSCs), thus limiting their application in tissue regeneration. The present study aimed to elucidate the role and mechanism of repetitive element (RE) activation in BMSC senescence and osteogenesis, as well as the intervention effect of quercetin. In an H2O2‑induced BMSC senescence model, quercetin treatment alleviated senescence as shown by a decrease in senescence‑associated β‑galactosidase (SA‑β‑gal)‑positive cell ratio, increased colony formation ability and decreased mRNA expression of p21 and senescence‑associated secretory phenotype genes. DNA damage response marker γ‑H2AX increased in senescent BMSCs, while expression of epigenetic markers methylation histone H3 Lys9, heterochromatin protein 1α and heterochromatin‑related nuclear membrane protein lamina‑associated polypeptide 2 decreased. Quercetin rescued these alterations, indicating its ability to ameliorate senescence by stabilizing heterochromatin structure where REs are primarily suppressed. Transcriptional activation of REs accompanied by accumulation of cytoplasmic double‑stranded (ds)RNA, as well as triggering of the RNA sensor retinoic acid‑inducible gene I (RIG‑I) receptor pathway in H2O2‑induced senescent BMSCs were shown. Similarly, quercetin treatment inhibited these responses. Additionally, RIG‑I knockdown led to a decreased number of SA‑β‑gal‑positive cells, confirming its functional impact on senescence. Induction of senescence or administration of dsRNA analogue significantly hindered the osteogenic capacity of BMSCs, while quercetin treatment or RIG‑I knockdown reversed the decline in osteogenic function. The findings of the current study demonstrated that quercetin inhibited the activation of REs and the RIG‑I RNA sensing pathway via epigenetic regulation, thereby alleviating the senescence of BMSCs and promoting osteogenesis.
Collapse
Affiliation(s)
- Yutong Sun
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510060, P.R. China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Chunyang Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510060, P.R. China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Liling Wen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510060, P.R. China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Zihang Ling
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510060, P.R. China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510060, P.R. China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510060, P.R. China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Jianmin Peng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510060, P.R. China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
12
|
Rigdon G, Prescott Y, Hall J, Abernathy K, Raskin J, Wargin W. Phase 1, Single-Center, Double-Blind, Randomized, Placebo-Controlled Studies of the Safety, Tolerability, and Pharmacokinetics of Single and Multiple Ascending Oral Doses of the Sirtuin 6 Activator SP-624 in Healthy Adults. Clin Pharmacol Drug Dev 2025; 14:18-25. [PMID: 39587867 PMCID: PMC11701958 DOI: 10.1002/cpdd.1488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
Sirtuin 6 activation is a novel epigenetic mechanism proposed for treatment of depression. Two Phase 1 studies, SP-624-101 and SP-624-102, examined the pharmacokinetics and safety of SP-624, an orally active sirtuin 6 activator, in healthy adults. SP-624-101 was a single-ascending-dose study. In Part A, participants were randomized 6:2 to SP-624 (single oral doses of 3, 10, or 30 mg) or placebo. Part B compared results in 8 participants receiving SP-624 while fasting or after a high-fat, high-calorie breakfast. In SP-624-102, a multiple-ascending-dose study, participants were randomized 6:2 to SP-624 (3 or 10 mg SP-624 daily) or placebo for 5 days and 5:2 to SP-624 20 mg daily or placebo for 10 days. At all doses, maximum concentration (Cmax) exceeded predicted target plasma concentrations of 3.28 ng/mL. Area under the concentration-time curve and Cmax increased dose proportionally. A food effect resulted in significantly lower Cmax, later time to maximum concentration, and comparable AUC for fed versus fasting participants. No serious adverse events were observed. In SP-624-101 and SP-624-102, respectively, 3 (12%) and 5 (29%) SP-624-treated participants experienced treatment-emergent adverse events. SP-624 was well tolerated and reached target concentrations in healthy adults, supporting progression of SP-624 20 mg daily into Phase 2 studies of major depressive disorder.
Collapse
Affiliation(s)
- Greg Rigdon
- Sirtsei Pharmaceuticals, Inc., a Subsidiary of Arrivo BioVenturesMorrisvilleNCUSA
| | - Yuki Prescott
- Sirtsei Pharmaceuticals, Inc., a Subsidiary of Arrivo BioVenturesMorrisvilleNCUSA
| | - John Hall
- Sirtsei Pharmaceuticals, Inc., a Subsidiary of Arrivo BioVenturesMorrisvilleNCUSA
| | - Kelly Abernathy
- Sirtsei Pharmaceuticals, Inc., a Subsidiary of Arrivo BioVenturesMorrisvilleNCUSA
| | - Joel Raskin
- Sirtsei Pharmaceuticals, Inc., a Subsidiary of Arrivo BioVenturesMorrisvilleNCUSA
| | - William Wargin
- Sirtsei Pharmaceuticals, Inc., a Subsidiary of Arrivo BioVenturesMorrisvilleNCUSA
| |
Collapse
|
13
|
Zhang Z, Huang H, Tao Y, Liu H, Fan Y. Sirt6 ameliorates high glucose-induced podocyte cytoskeleton remodeling via the PI3K/AKT signaling pathway. Ren Fail 2024; 46:2410396. [PMID: 39378103 PMCID: PMC11463017 DOI: 10.1080/0886022x.2024.2410396] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Podocyte injury plays an important role in the occurrence and progression of diabetic kidney disease (DKD), which leads to albuminuria. Cytoskeletal remodeling is an early manifestation of podocyte injury in DKD. However, the underlying mechanism of cytoskeletal remodeling has not been clarified. Histone deacetylase sirtuin6 (Sirt6) has been found to play a key role in DKD progression, and the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/AKT) pathway directly regulates the cytoskeletal structure of podocytes. Whereas, the relationship between Sirt6, the PI3K/AKT pathway and DKD progression remains unclear. METHODS Renal injury of db/db mice was observed by PAS staining and transmission electron microscope. Expression of Sirt6 in the glomeruli of db/db mice was detected by immunofluorescence. UBCS039, a Sirt6 activator, was used to explore the renal effects of Sirt6 activation on diabetic mouse kidneys. We also downregulating Sirt6 expression in podocytes using the Sirt6 inhibitor, OSS_128167, and induced upregulation of Sirt6 using a recombinant plasmid, after which the effects of Sirt6 on high glucose (HG)-induced podocyte damage were assessed in vitro. Podocyte cytoskeletal structures were observed by phalloidin staining. The podocyte apoptotic rate was assessed by flow cytometry, and PI3K/AKT signaling activation was measured by Western blotting. RESULTS Db/db mice exhibited renal damage including elevated urine albumin-to-creatinine ratio (ACR), increased mesangial matrix, fused podocyte foot processes, and thickened glomerular basement membrane. The expression of Sirt6 and PI3K/AKT pathway components was decreased in db/db mice. UBCS039 increased the expressions of Sirt6 and PI3K/AKT pathway components and ameliorated renal damage in db/db mice. We also observed consistent Sirt6 expression was in HG-induced podocytes in vitro. Activation of the PI3K/AKT pathway via a Sirt6 recombinant plasmid ameliorated podocyte cytoskeletal remodeling and apoptosis in HG-treated immortalized human podocytes in vitro, whereas Sirt6 inhibition by OSS_128167 accelerated HG-induced podocyte damage in vitro. CONCLUSIONS Sirt6 protects podocytes against HG-induced cytoskeletal remodeling and apoptosis through activation of the PI3K/AKT signaling pathway. These findings provide evidence supporting the potential efficacy of Sirt6 activation as a promising therapeutic strategy for addressing podocyte injury in DKD.
Collapse
Affiliation(s)
- Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hao Huang
- Division of Rehabilitation, Tianmen First People’s Hospital, Tianmen, Hubei, China
| | - Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, USA
| | - Hongyan Liu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yanqin Fan
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
14
|
Du J, Yin Y, Wu D, Diao C, Zhao T, Peng F, Li N, Wang D, Shi J, Wang L, Kong L, Zhou W, Hao A. SIRT6 modulates lesion microenvironment in LPC induced demyelination by targeting astrocytic CHI3L1. J Neuroinflammation 2024; 21:243. [PMID: 39342313 PMCID: PMC11438192 DOI: 10.1186/s12974-024-03241-1] [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: 06/17/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
Demyelination occurs widely in the central nervous system (CNS) neurodegenerative diseases, especially the multiple sclerosis (MS), which with a complex and inflammatory lesion microenvironment inhibiting remyelination. Sirtuin6 (SIRT6), a histone/protein deacetylase is of interest for its promising effect in transcriptional regulation, cell cycling, inflammation, metabolism and longevity. Here we show that SIRT6 participates in the remyelination process in mice subjected to LPC-induced demyelination. Using pharmacological SIRT6 inhibitor or activator, we found that SIRT6 modulated LPC-induced damage in motor or cognitive function. Inhibition of SIRT6 impaired myelin regeneration, exacerbated neurological deficits, and decreased oligodendrocyte precursor cells (OPCs) proliferation and differentiation, whereas activation of SIRT6 reversed behavioral performance in mice, demonstrating a beneficial effect of SIRT6. Importantly, based on RNA sequencing analysis of the corpus callosum tissues, it was further revealed that SIRT6 took charge in regulation of glial activation during remyelination, and significant alterations in CHI3L1 were obtained, a glycoprotein specifically secreted by astrocytes. Impaired proliferation and differentiation of OPCs could be induced in vitro using supernatants from reactive astrocyte, especially when SIRT6 was inhibited. Mechanistically, SIRT6 regulates the secretion of CHI3L1 from reactive astrocytes by histone-H3-lysine-9 acetylation (H3K9Ac). Adeno-associated virus-overexpression of SIRT6 (AAV-SIRT6-OE) in astrocytes improved remyelination and functional recovery after LPC-induced demyelination, whereas together with AAV-CHI3L1-OE inhibits this therapeutic effect. Collectively, our data elucidate the role of SIRT6 in remyelination and further reveal astrocytic SIRT6/CHI3L1 as the key regulator for improving the remyelination environment, which may be a potential target for MS therapy.
Collapse
Affiliation(s)
- Jingyi Du
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yue Yin
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Dong Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Can Diao
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Tiantian Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Fan Peng
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Naigang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Dongshuang Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Jiaming Shi
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Liyan Wang
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjuan Zhou
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China.
| | - Aijun Hao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
15
|
Bellani MA, Shaik A, Majumdar I, Ling C, Seidman MM. Repair of genomic interstrand crosslinks. DNA Repair (Amst) 2024; 141:103739. [PMID: 39106540 PMCID: PMC11423799 DOI: 10.1016/j.dnarep.2024.103739] [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: 03/12/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024]
Abstract
Genomic interstrand crosslinks (ICLs) are formed by reactive species generated during normal cellular metabolism, produced by the microbiome, and employed in cancer chemotherapy. While there are multiple options for replication dependent and independent ICL repair, the crucial step for each is unhooking one DNA strand from the other. Much of our insight into mechanisms of unhooking comes from powerful model systems based on plasmids with defined ICLs introduced into cells or cell free extracts. Here we describe the properties of exogenous and endogenous ICL forming compounds and provide an historical perspective on early work on ICL repair. We discuss the modes of unhooking elucidated in the model systems, the concordance or lack thereof in drug resistant tumors, and the evolving view of DNA adducts, including ICLs, formed by metabolic aldehydes.
Collapse
Affiliation(s)
- Marina A Bellani
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Althaf Shaik
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ishani Majumdar
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Chen Ling
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael M Seidman
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
16
|
Zhang Z, Jiang Z, Cheng J, Price CA, Yang L, Li Q. Nicotine induces senescence in spermatogonia stem cells by disrupting homeostasis between circadian oscillation and rhythmic mitochondrial dynamics via the SIRT6/Bmal1 pathway. Life Sci 2024; 352:122860. [PMID: 38936603 DOI: 10.1016/j.lfs.2024.122860] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/15/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Infertility is intricately linked with alterations in circadian rhythms along with physiological decline and stem cell senescence. Yet, the direct involvement of circadian mechanisms in nicotine-induced injury to the testes, especially the senescence of spermatogonia stem cells (SSCs), is not well comprehended. This study revealed that nicotine exposure induced testis injury by triggering SSCs senescence along with the upregulation of senescence marker genes and senescence-associated secretory phenotype components. Moreover, nicotine treatment caused mitochondrial hyper-fusion, increased oxidative stress, and DNA damage. Exposure to nicotine was found to suppress the expression of sirtuin 6 (SIRT6), which accelerated the senescence of spermatogonia stem cells (SSCs). This acceleration led to increased acetylation of brain and muscle ARNT-like protein (Bmal1), consequently reducing the expression of Bmal1 protein. Conversely, the overexpression of Bmal1 alleviated mitochondrial hyper-fusion and senescence phenotypes induced by nicotine. Overall, this study unveiled a novel molecular mechanism behind nicotine-induced disorders in spermatogenesis and highlighted the SIRT6/Bmal1 regulatory pathway as a potential therapeutic target for combating nicotine-associated infertility.
Collapse
Affiliation(s)
- Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhongliang Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jianyong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Christopher A Price
- Centre de recherche en reproduction & fertility, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 7C6, Canada
| | - Li Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
17
|
Buriánek F, Gege C, Marinković P. New developments in celiac disease treatments. Drug Discov Today 2024; 29:104113. [PMID: 39067614 DOI: 10.1016/j.drudis.2024.104113] [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: 02/19/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Celiac disease (CeD), an autoimmune disorder triggered by gluten, affects around 1% of the global population. Standard treatment is a strict gluten-free diet (GFD), which poses significant challenges due to dietary restrictions, cross-contamination and subsequent persistent intestinal inflammation. This underscores the need for new treatment options addressing the complex pathophysiology of CeD. Recent research focuses on developing drugs that target intestinal barrier regeneration, gluten peptide modification, immune response alteration, and gut microbial ecosystem modulation. These approaches offer potential for more effective management of CeD beyond GFD. Gluten-independent treatments may be particularly relevant under the FDA's draft guidance for CeD, which emphasizes drug development as an adjunct to GFD for patients with ongoing signs and symptoms of CeD despite strict GFD.
Collapse
|
18
|
Frohlich J, Liorni N, Mangoni M, Lochmanová G, Pírek P, Kaštánková N, Pata P, Kucera J, Chaldakov GN, Tonchev AB, Pata I, Gorbunova V, Leire E, Zdráhal Z, Mazza T, Vinciguerra M. Epigenetic and transcriptional control of adipocyte function by centenarian-associated SIRT6 N308K/A313S mutant. Clin Epigenetics 2024; 16:96. [PMID: 39033117 PMCID: PMC11265064 DOI: 10.1186/s13148-024-01710-1] [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: 05/14/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Obesity is a major health burden. Preadipocytes proliferate and differentiate in mature adipocytes in the adipogenic process, which could be a potential therapeutic approach for obesity. Deficiency of SIRT6, a stress-responsive protein deacetylase and mono-ADP ribosyltransferase enzyme, blocks adipogenesis. Mutants of SIRT6 (N308K/A313S) were recently linked to the in the long lifespan Ashkenazi Jews. In this study, we aimed to clarify how these new centenarian-associated SIRT6 genetic variants affect adipogenesis at the transcriptional and epigenetic level. METHODS We analyzed the role of SIRT6 wild-type (WT) or SIRT6 centenarian-associated mutant (N308K/A313S) overexpression in adipogenesis, by creating stably transduced preadipocyte cell lines using lentivirus on the 3T3-L1 model. Histone post-translational modifications (PTM: acetylation, methylation) and transcriptomic changes were analyzed by mass spectrometry (LC-MS/MS) and RNA-Seq, respectively, in 3T3-L1 adipocytes. In addition, the adipogenic process and related signaling pathways were investigated by bioinformatics and biochemical approaches. RESULTS Overexpression of centenarian-associated SIRT6 mutant increased adipogenic differentiation to a similar extent compared to the WT form. However, it triggered distinct histone PTM profiles in mature adipocytes, with significantly higher acetylation levels, and activated divergent transcriptional programs, including those dependent on signaling related to the sympathetic innervation and to PI3K pathway. 3T3-L1 mature adipocytes overexpressing SIRT6 N308K/A313S displayed increased insulin sensitivity in a neuropeptide Y (NPY)-dependent manner. CONCLUSIONS SIRT6 N308K/A313S overexpression in mature adipocytes ameliorated glucose sensitivity and impacted sympathetic innervation signaling. These findings highlight the importance of targeting SIRT6 enzymatic activities to regulate the co-morbidities associated with obesity.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Niccolò Liorni
- IRCCS, Bioinformatics Unit, Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Manuel Mangoni
- IRCCS, Bioinformatics Unit, Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Gabriela Lochmanová
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Laboratory of Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Pavlína Pírek
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Nikola Kaštánková
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | | | - Jan Kucera
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Physical Activities and Health, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| | - George N Chaldakov
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Faculty of Medicine, Varna, Bulgaria
| | - Anton B Tonchev
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Faculty of Medicine, Varna, Bulgaria
| | | | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Eric Leire
- GenFlow Biosciences Srl, Charleroi, Belgium
- Clinique 135, Brussels, Belgium
| | - Zbyněk Zdráhal
- Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Laboratory of Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Tommaso Mazza
- IRCCS, Bioinformatics Unit, Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic.
- Department of Translational Stem Cell Biology, Research Institute of the Medical University, Varna, Bulgaria.
- Faculty of Science, Liverpool John Moores University (LJMU), Liverpool, UK.
| |
Collapse
|
19
|
Arellano MYG, VanHeest M, Emmadi S, Abdul-Hafez A, Ibrahim SA, Thiruvenkataramani RP, Teleb RS, Omar H, Kesaraju T, Mohamed T, Madhukar BV, Omar SA. Role of Mesenchymal Stem/Stromal Cells (MSCs) and MSC-Derived Extracellular Vesicles (EVs) in Prevention of Telomere Length Shortening, Cellular Senescence, and Accelerated Biological Aging. Bioengineering (Basel) 2024; 11:524. [PMID: 38927760 PMCID: PMC11200821 DOI: 10.3390/bioengineering11060524] [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: 03/19/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Biological aging is defined as a progressive decline in tissue function that eventually results in cell death. Accelerated biologic aging results when the telomere length is shortened prematurely secondary to damage from biological or environmental stressors, leading to a defective reparative mechanism. Stem cells therapy may have a potential role in influencing (counteract/ameliorate) biological aging and maintaining the function of the organism. Mesenchymal stem cells, also called mesenchymal stromal cells (MSCs) are multipotent stem cells of mesodermal origin that can differentiate into other types of cells, such as adipocytes, chondrocytes, and osteocytes. MSCs influence resident cells through the secretion of paracrine bioactive components such as cytokines and extracellular vesicles (EVs). This review examines the changes in telomere length, cellular senescence, and normal biological age, as well as the factors contributing to telomere shortening and accelerated biological aging. The role of MSCs-especially those derived from gestational tissues-in prevention of telomere shortening (TS) and accelerated biological aging is explored. In addition, the strategies to prevent MSC senescence and improve the antiaging therapeutic application of MSCs and MSC-derived EVs in influencing telomere length and cellular senescence are reviewed.
Collapse
Affiliation(s)
- Myrna Y. Gonzalez Arellano
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Matthew VanHeest
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sravya Emmadi
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Amal Abdul-Hafez
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Sherif Abdelfattah Ibrahim
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ranga P. Thiruvenkataramani
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Rasha S. Teleb
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Department of Pediatrics and Neonatology, Qena Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Hady Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tulasi Kesaraju
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
| | - Tarek Mohamed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| | - Burra V. Madhukar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
| | - Said A. Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.Y.G.A.); (A.A.-H.); (S.A.I.); (R.P.T.); (R.S.T.); (H.O.); (T.K.); (T.M.); (B.V.M.)
- College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (M.V.); (S.E.)
- Regional Neonatal Intensive Care Unit, Sparrow Hospital, Lansing, MI 48912, USA
| |
Collapse
|
20
|
Jia S, Liang R, Chen J, Liao S, Lin J, Li W. Emerging technology has a brilliant future: the CRISPR-Cas system for senescence, inflammation, and cartilage repair in osteoarthritis. Cell Mol Biol Lett 2024; 29:64. [PMID: 38698311 PMCID: PMC11067114 DOI: 10.1186/s11658-024-00581-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Osteoarthritis (OA), known as one of the most common types of aseptic inflammation of the musculoskeletal system, is characterized by chronic pain and whole-joint lesions. With cellular and molecular changes including senescence, inflammatory alterations, and subsequent cartilage defects, OA eventually leads to a series of adverse outcomes such as pain and disability. CRISPR-Cas-related technology has been proposed and explored as a gene therapy, offering potential gene-editing tools that are in the spotlight. Considering the genetic and multigene regulatory mechanisms of OA, we systematically review current studies on CRISPR-Cas technology for improving OA in terms of senescence, inflammation, and cartilage damage and summarize various strategies for delivering CRISPR products, hoping to provide a new perspective for the treatment of OA by taking advantage of CRISPR technology.
Collapse
Affiliation(s)
- Shicheng Jia
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Rongji Liang
- Shantou University Medical College, Shantou, 515041, China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Shuai Liao
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Wei Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
21
|
Dhiman S, Mannan A, Taneja A, Mohan M, Singh TG. Sirtuin dysregulation in Parkinson's disease: Implications of acetylation and deacetylation processes. Life Sci 2024; 342:122537. [PMID: 38428569 DOI: 10.1016/j.lfs.2024.122537] [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: 01/03/2024] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative condition that primarily affects motor function and is caused by a gradual decline of dopaminergic neurons in the brain's substantia pars compacta (Snpc) region. Multiple molecular pathways are involved in the pathogenesis, which results in impaired cellular functions and neuronal degeneration. However, the role of sirtuins, a type of NAD+-dependent deacetylase, in the pathogenesis of Parkinson's disease has recently been investigated. Sirtuins are essential for preserving cellular homeostasis because they control a number of biological processes, such as metabolism, apoptosis, and DNA repair. This review shed lights on the dysregulation of sirtuin activity in PD, highlighting the role that acetylation and deacetylation processes play in the development of the disease. Key regulators of protein acetylation, sirtuins have been found to be involved in the aberrant acetylation of vital substrates linked to PD pathology when their balance is out of balance. The hallmark characteristics of PD such as neuroinflammation, oxidative stress, and mitochondrial dysfunction have all been linked to the dysregulation of sirtuin expression and activity. Furthermore, we have also explored how the modulators of sirtuins can be a promising therapeutic intervention in the treatment of PD.
Collapse
Affiliation(s)
- Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Ayushi Taneja
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
22
|
Al-Danakh A, Safi M, Jian Y, Yang L, Zhu X, Chen Q, Yang K, Wang S, Zhang J, Yang D. Aging-related biomarker discovery in the era of immune checkpoint inhibitors for cancer patients. Front Immunol 2024; 15:1348189. [PMID: 38590525 PMCID: PMC11000233 DOI: 10.3389/fimmu.2024.1348189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/29/2024] [Indexed: 04/10/2024] Open
Abstract
Older patients with cancer, particularly those over 75 years of age, often experience poorer clinical outcomes compared to younger patients. This can be attributed to age-related comorbidities, weakened immune function, and reduced tolerance to treatment-related adverse effects. In the immune checkpoint inhibitors (ICI) era, age has emerged as an influential factor impacting the discovery of predictive biomarkers for ICI treatment. These age-linked changes in the immune system can influence the composition and functionality of tumor-infiltrating immune cells (TIICs) that play a crucial role in the cancer response. Older patients may have lower levels of TIICs infiltration due to age-related immune senescence particularly T cell function, which can limit the effectivity of cancer immunotherapies. Furthermore, age-related immune dysregulation increases the exhaustion of immune cells, characterized by the dysregulation of ICI-related biomarkers and a dampened response to ICI. Our review aims to provide a comprehensive understanding of the mechanisms that contribute to the impact of age on ICI-related biomarkers and ICI response. Understanding these mechanisms will facilitate the development of treatment approaches tailored to elderly individuals with cancer.
Collapse
Affiliation(s)
- Abdullah Al-Danakh
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mohammed Safi
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yuli Jian
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Linlin Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xinqing Zhu
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qiwei Chen
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Kangkang Yang
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Deyong Yang
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Surgery, Healinghands Clinic, Dalian, Liaoning, China
| |
Collapse
|
23
|
Xu X, Zhang Q, Wang X, Jin J, Wu C, Feng L, Yang X, Zhao M, Chen Y, Lu S, Zheng Z, Lan X, Wang Y, Zheng Y, Lu X, Zhang Q, Zhang J. Discovery of a potent and highly selective inhibitor of SIRT6 against pancreatic cancer metastasis in vivo. Acta Pharm Sin B 2024; 14:1302-1316. [PMID: 38487000 PMCID: PMC10935062 DOI: 10.1016/j.apsb.2023.11.014] [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: 07/17/2023] [Revised: 09/05/2023] [Accepted: 10/18/2023] [Indexed: 03/17/2024] Open
Abstract
Pancreatic cancer, one of the most aggressive malignancies, has no effective treatment due to the lack of targets and drugs related to tumour metastasis. SIRT6 can promote the migration of pancreatic cancer and could be a potential target for antimetastasis of pancreatic cancer. However, highly selective and potency SIRT6 inhibitor that can be used in vivo is yet to be discovered. Here, we developed a novel SIRT6 allosteric inhibitor, compound 11e, with maximal inhibitory potency and an IC50 value of 0.98 ± 0.13 μmol/L. Moreover, compound 11e exhibited significant selectivity against other histone deacetylases (HADC1‒11 and SIRT1‒3) at concentrations up to 100 μmol/L. The allosteric site and the molecular mechanism of inhibition were extensively elucidated by cocrystal complex structure and dynamic structural analyses. Importantly, we confirmed the antimetastatic function of such inhibitors in four pancreatic cancer cell lines as well as in two mouse models of pancreatic cancer liver metastasis. To our knowledge, this is the first study to reveal the in vivo effects of SIRT6 inhibitors on liver metastatic pancreatic cancer. It not only provides a promising lead compound for subsequent inhibitor development targeting SIRT6 but also provides a potential approach to address the challenge of metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Xinyuan Xu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and BioinformaticsCenter, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Zhang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and BioinformaticsCenter, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xufeng Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Jing Jin
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230026, China
| | - Chengwei Wu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and BioinformaticsCenter, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Feng
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and BioinformaticsCenter, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiuyan Yang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mingzhu Zhao
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yingyi Chen
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shaoyong Lu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhen Zheng
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaobing Lan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Yi Wang
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei 230026, China
| | - Yan Zheng
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xuefeng Lu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qiufen Zhang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Zhang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Medicinal Chemistry and BioinformaticsCenter, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
24
|
Zheng L, He S, Wang H, Li J, Liu Y, Liu S. Targeting Cellular Senescence in Aging and Age-Related Diseases: Challenges, Considerations, and the Emerging Role of Senolytic and Senomorphic Therapies. Aging Dis 2024; 15:2554-2594. [PMID: 38421832 PMCID: PMC11567261 DOI: 10.14336/ad.2024.0206] [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: 12/12/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Cellular senescence is characterized by the permanent arrest of cell proliferation and is a response to endogenous and exogenous stress. The continuous accumulation of senescent cells (SnCs) in the body leads to the development of aging and age-related diseases (such as neurodegenerative diseases, cancer, metabolic diseases, cardiovascular diseases, and osteoarthritis). In the face of the growing challenge of aging and age-related diseases, several compounds have received widespread attention for their potential to target SnCs. As a result, senolytics (compounds that selectively eliminate SnCs) and senomorphics (compounds that alter intercellular communication and modulate the behavior of SnCs) have become hot research topics in the field of anti-aging. In addition, strategies such as combination therapies and immune-based approaches have also made significant progress in the field of anti-aging therapy. In this article, we discuss the latest research on anti-aging targeting SnCs and gain a deeper understanding of the mechanism of action and impact of different anti-aging strategies on aging and age-related diseases, with the aim of providing more effective references and therapeutic ideas for clinical anti-aging treatment in the face of the ever-grave challenges of aging and age-related diseases.
Collapse
Affiliation(s)
- Liyao Zheng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shipei He
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuanyuan Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China.
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
25
|
Shalwitz R, Day T, Ruehlmann AK, Julio L, Gordon S, Vandeuren A, Nelson M, Lyman M, Kelly K, Altvater A, Ondeck C, O'Brien S, Hamilton T, Hanson RL, Wayman K, Miller A, Shalwitz I, Batchelor E, McNutt P. Treatment of Sulfur Mustard Corneal Injury by Augmenting the DNA Damage Response (DDR): A Novel Approach. J Pharmacol Exp Ther 2024; 388:526-535. [PMID: 37977813 PMCID: PMC10801765 DOI: 10.1124/jpet.123.001686] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 11/19/2023] Open
Abstract
Sulfur mustard (SM) is a highly reactive organic chemical has been used as a chemical warfare agent and terrorist threat since World War I. The cornea is highly sensitive to SM toxicity and exposure to low vapor doses can cause incapacitating acute injuries. Exposure to higher doses can elicit persistent secondary keratopathies that cause reduced quality of life and impaired or lost vision. Despite a century of research, there are no specific treatments for acute or persistent ocular SM injuries. SM cytotoxicity emerges, in part, through DNA alkylation and double-strand breaks (DSBs). Because DSBs can naturally be repaired by DNA damage response pathways with low efficiency, we hypothesized that enhancing the homologous recombination pathway could pose a novel approach to mitigate SM injury. Here, we demonstrate that a dilithium salt of adenosine diphosphoribose (INV-102) increases protein levels of p53 and Sirtuin 6, upregulates transcription of BRCA1/2, enhances γH2AX focus formation, and promotes assembly of repair complexes at DSBs. Based on in vitro evidence showing INV-102 enhancement of DNA damage response through both p53-dependent and p53-independent pathways, we next tested INV-102 in a rabbit preclinical model of corneal injury. In vivo studies demonstrate a marked reduction in the incidence and severity of secondary keratopathies in INV-102-treated eyes compared with vehicle-treated eyes when treatment was started 24 hours after SM vapor exposure. These results suggest DNA repair mechanisms are a viable therapeutic target for SM injury and suggest topical treatment with INV-102 is a promising approach for SM as well as other conditions associated with DSBs. SIGNIFICANCE STATEMENT: Sulfur mustard gas corneal injury currently has no therapeutic treatment. This study aims to show the therapeutic potential of activating the body's natural DNA damage response to activate tissue repair.
Collapse
Affiliation(s)
- Robert Shalwitz
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Tovah Day
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Anna Kotsakis Ruehlmann
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Lindsay Julio
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Shellaina Gordon
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Adrianna Vandeuren
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Marian Nelson
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Megan Lyman
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Kyle Kelly
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Amber Altvater
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Celinia Ondeck
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Sean O'Brien
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Tracey Hamilton
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Ryan L Hanson
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Kayla Wayman
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Alexandrea Miller
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Isaiah Shalwitz
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Eric Batchelor
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| | - Patrick McNutt
- Invirsa, Inc., Columbus, Ohio (R.S., A.K.R., A.M., I.S.); Department of Biology, Northeastern University, Boston, Massachusetts (T.D., L.J., S.G., A.V.); Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota (R.L.H., K.W., E.B.); United States Army Medical Research Institute for Chemical Defense, Gunpowder, Maryland (M.N., M.L., K.K., A.A., C.O., S.O., T.H., P.M.); and Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina (S.O., C.O., P.M.)
| |
Collapse
|
26
|
Chini CCS, Cordeiro HS, Tran NLK, Chini EN. NAD metabolism: Role in senescence regulation and aging. Aging Cell 2024; 23:e13920. [PMID: 37424179 PMCID: PMC10776128 DOI: 10.1111/acel.13920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
The geroscience hypothesis proposes that addressing the biology of aging could directly prevent the onset or mitigate the severity of multiple chronic diseases. Understanding the interplay between key aspects of the biological hallmarks of aging is essential in delivering the promises of the geroscience hypothesis. Notably, the nucleotide nicotinamide adenine dinucleotide (NAD) interfaces with several biological hallmarks of aging, including cellular senescence, and changes in NAD metabolism have been shown to be involved in the aging process. The relationship between NAD metabolism and cellular senescence appears to be complex. On the one hand, the accumulation of DNA damage and mitochondrial dysfunction induced by low NAD+ can promote the development of senescence. On the other hand, the low NAD+ state that occurs during aging may inhibit SASP development as this secretory phenotype and the development of cellular senescence are both highly metabolically demanding. However, to date, the impact of NAD+ metabolism on the progression of the cellular senescence phenotype has not been fully characterized. Therefore, to explore the implications of NAD metabolism and NAD replacement therapies, it is essential to consider their interactions with other hallmarks of aging, including cellular senescence. We propose that a comprehensive understanding of the interplay between NAD boosting strategies and senolytic agents is necessary to advance the field.
Collapse
Affiliation(s)
- Claudia Christiano Silva Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Heidi Soares Cordeiro
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Ngan Le Kim Tran
- Center for Clinical and Translational Science and Mayo Clinic Graduate School of Biomedical SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Eduardo Nunes Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| |
Collapse
|
27
|
Minisini M, Cricchi E, Brancolini C. Acetylation and Phosphorylation in the Regulation of Hypoxia-Inducible Factor Activities: Additional Options to Modulate Adaptations to Changes in Oxygen Levels. Life (Basel) 2023; 14:20. [PMID: 38276269 PMCID: PMC10821055 DOI: 10.3390/life14010020] [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: 12/05/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
O2 is essential for the life of eukaryotic cells. The ability to sense oxygen availability and initiate a response to adapt the cell to changes in O2 levels is a fundamental achievement of evolution. The key switch for adaptation consists of the transcription factors HIF1A, HIF2A and HIF3A. Their levels are tightly controlled by O2 through the involvement of the oxygen-dependent prolyl hydroxylase domain-containing enzymes (PHDs/EGNLs), the von Hippel-Lindau tumour suppressor protein (pVHL) and the ubiquitin-proteasome system. Furthermore, HIF1A and HIF2A are also under the control of additional post-translational modifications (PTMs) that positively or negatively regulate the activities of these transcription factors. This review focuses mainly on two PTMs of HIF1A and HIF2A: phosphorylation and acetylation.
Collapse
Affiliation(s)
| | | | - Claudio Brancolini
- Lab of Epigenomics, Department of Medicine, Università degli Studi di Udine, 33100 Udine, Italy; (M.M.); (E.C.)
| |
Collapse
|
28
|
Cheng J, Keuthan CJ, Esumi N. The many faces of SIRT6 in the retina and retinal pigment epithelium. Front Cell Dev Biol 2023; 11:1244765. [PMID: 38016059 PMCID: PMC10646311 DOI: 10.3389/fcell.2023.1244765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/18/2023] [Indexed: 11/30/2023] Open
Abstract
Sirtuin 6 (SIRT6) is a member of the mammalian sirtuin family of NAD+-dependent protein deacylases, homologues of the yeast silent information regulator 2 (Sir2). SIRT6 has remarkably diverse functions and plays a key role in a variety of biological processes for maintaining cellular and organismal homeostasis. In this review, our primary aim is to summarize recent progress in understanding SIRT6's functions in the retina and retinal pigment epithelium (RPE), with the hope of further drawing interests in SIRT6 to increase efforts in exploring the therapeutic potential of this unique protein in the vision field. Before describing SIRT6's role in the eye, we first discuss SIRT6's general functions in a wide range of biological contexts. SIRT6 plays an important role in gene silencing, metabolism, DNA repair, antioxidant defense, inflammation, aging and longevity, early development, and stress response. In addition, recent studies have revealed SIRT6's role in macrophage polarization and mitochondrial homeostasis. Despite being initially understudied in the context of the eye, recent efforts have begun to elucidate the critical functions of SIRT6 in the retina and RPE. In the retina, SIRT6 is essential for adult retinal function, regulates energy metabolism by suppressing glycolysis that affects photoreceptor cell survival, protects retinal ganglion cells from oxidative stress, and plays a role in Müller cells during early neurodegenerative events in diabetic retinopathy. In the RPE, SIRT6 activates autophagy in culture and protects against oxidative stress in mice. Taken together, this review demonstrates that better understanding of SIRT6's functions and their mechanisms, both in and out of the context of the eye, holds great promise for the development of SIRT6-targeted strategies for prevention and treatment of blinding eye diseases.
Collapse
Affiliation(s)
| | | | - Noriko Esumi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
29
|
Xu Y, Xiao W. NAD+: An Old but Promising Therapeutic Agent for Skeletal Muscle Ageing. Ageing Res Rev 2023; 92:102106. [PMID: 39492424 DOI: 10.1016/j.arr.2023.102106] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2024]
Abstract
More than a century after the discovery of nicotinamide adenine dinucleotide (NAD+), our understanding of the molecule's role in the biology of ageing continues to evolve. As a coenzyme or substrate for many enzymes, NAD+ governs a wide range of biological processes, including energy metabolism, genomic stability, signal transduction, and cell fate. NAD+ deficiency has been recognised as a bona fide hallmark of tissue degeneration, and restoring NAD+ homeostasis helps to rejuvenate multiple mechanisms associated with tissue ageing. The progressive loss of skeletal muscle homeostasis with age is directly associated with high morbidity, disability and mortality. The aetiology of skeletal muscle ageing is complex, involving mitochondrial dysfunction, senescence and stem cell depletion, autophagy defects, chronic cellular stress, intracellular ion overload, immune cell dysfunction, circadian clock disruption, microcirculation disorders, persistent denervation, and gut microbiota dysbiosis. This review focuses on the therapeutic potential of NAD+ restoration to alleviate the above pathological factors and discusses the effects of in vivo administration of different NAD+ boosting strategies on skeletal muscle homeostasis, aiming to provide a reference for combating skeletal muscle ageing.
Collapse
Affiliation(s)
- Yingying Xu
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Weihua Xiao
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
30
|
Zhang M, Liu W, Liu Y, Zhang Z, Hu Y, Sun D, Li S, Fang J. Astragaloside IV Inhibited Podocyte Pyroptosis in Diabetic Kidney Disease by Regulating SIRT6/HIF-1α Axis. DNA Cell Biol 2023; 42:594-607. [PMID: 37751175 DOI: 10.1089/dna.2023.0102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
To investigate the effect of astragaloside IV (AS) on podocytes pyroptosis in diabetic kidney disease (DKD). Forty male Sprague-Dawley rats were randomly divided into normal group (n = 10) and model group (n = 30). Rats in model group were intraperitoneally injected streptozotocin (60 mg/kg) for 3 days to induce DKD. Then rats were divided into DKD group, AS group, and UBCS group. The AS group was given 40 mg/kg/d of AS by gavage, and UBCS group was given 50 mg/kg/d of UBCS039 by gavage, and normal group and DKD group were given the same amount saline for 8 weeks, once a day. Hematoxylin-eosin and masson staining were used to observe pathology of kidney. Rat podocytes were divided into normal group, mannitol hypertonic group, high-glucose group, UBCS group, OSS group, and AS group. Western blotting, quantitative real-time polymerase chain reaction, immunofluorescence, and flow cytometry were used to analyze pyroptosis-related markers and reactive oxygen species (ROS) levels. Results showed that AS inhibited ROS and alleviated podocytes pyroptosis in rats by increasing expression of sirtuin 6 (SIRT6) and decreasing expression of hypoxia inducible factor 1 subunit alpha (HIF-1α). UBCS039 and AS enhanced SIRT6 level, decreased HIF-1α level, and finally improved pyroptosis of podocytes in vitro, whereas OSS-128167 showed the opposite effect for podocytes pyroptosis. AS improved podocytes pyroptosis in DKD by regulating SIRT6/HIF-1α pathway, thereby alleviating injury of DKD.
Collapse
Affiliation(s)
- Mingyu Zhang
- Inner Mongolia Baogang Hospital, Baotou, China
- Department of Nephrology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | | | | | | | - Yaling Hu
- Shanxi Medical University, Taiyuan, China
| | - Dalin Sun
- Inner Mongolia Baogang Hospital, Baotou, China
| | - Sufen Li
- Shanxi Medical University, Taiyuan, China
| | | |
Collapse
|
31
|
Xiao H, Xie Y, Xi K, Xie J, Liu M, Zhang Y, Cheng Z, Wang W, Guo B, Wu S. Targeting Mitochondrial Sirtuins in Age-Related Neurodegenerative Diseases and Fibrosis. Aging Dis 2023; 14:1583-1605. [PMID: 37196115 PMCID: PMC10529758 DOI: 10.14336/ad.2023.0203] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/03/2023] [Indexed: 05/19/2023] Open
Abstract
Aging is a natural and complex biological process that is associated with widespread functional declines in numerous physiological processes, terminally affecting multiple organs and tissues. Fibrosis and neurodegenerative diseases (NDs) often occur with aging, imposing large burdens on public health worldwide, and there are currently no effective treatment strategies for these diseases. Mitochondrial sirtuins (SIRT3-5), which are members of the sirtuin family of NAD+-dependent deacylases and ADP-ribosyltransferases, are capable of regulating mitochondrial function by modifying mitochondrial proteins that participate in the regulation of cell survival under various physiological and pathological conditions. A growing body of evidence has revealed that SIRT3-5 exert protective effects against fibrosis in multiple organs and tissues, including the heart, liver, and kidney. SIRT3-5 are also involved in multiple age-related NDs, including Alzheimer's disease, Parkinson's disease, and Huntington's disease. Furthermore, SIRT3-5 have been noted as promising targets for antifibrotic therapies and the treatment of NDs. This review systematically highlights recent advances in knowledge regarding the role of SIRT3-5 in fibrosis and NDs and discusses SIRT3-5 as therapeutic targets for NDs and fibrosis.
Collapse
Affiliation(s)
- Haoxiang Xiao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Yuqiao Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Kaiwen Xi
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Jinyi Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Mingyue Liu
- Medical School, Yan’an University, Yan’an, China
| | - Yangming Zhang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Zishuo Cheng
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Baolin Guo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China.
| |
Collapse
|
32
|
Ye Y, Yang K, Liu H, Yu Y, Song M, Huang D, Lei J, Zhang Y, Liu Z, Chu Q, Fan Y, Zhang S, Jing Y, Esteban CR, Wang S, Belmonte JCI, Qu J, Zhang W, Liu GH. SIRT2 counteracts primate cardiac aging via deacetylation of STAT3 that silences CDKN2B. NATURE AGING 2023; 3:1269-1287. [PMID: 37783815 DOI: 10.1038/s43587-023-00486-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 08/15/2023] [Indexed: 10/04/2023]
Abstract
Aging is a major risk factor contributing to pathophysiological changes in the heart, yet its intrinsic mechanisms have been largely unexplored in primates. In this study, we investigated the hypertrophic and senescence phenotypes in the hearts of aged cynomolgus monkeys as well as the transcriptomic and proteomic landscapes of young and aged primate hearts. SIRT2 was identified as a key protein decreased in aged monkey hearts, and engineered SIRT2 deficiency in human pluripotent stem cell-derived cardiomyocytes recapitulated key senescence features of primate heart aging. Further investigations revealed that loss of SIRT2 in human cardiomyocytes led to the hyperacetylation of STAT3, which transcriptionally activated CDKN2B and, in turn, triggered cardiomyocyte degeneration. Intra-myocardial injection of lentiviruses expressing SIRT2 ameliorated age-related cardiac dysfunction in mice. Taken together, our study provides valuable resources for decoding primate cardiac aging and identifies the SIRT2-STAT3-CDKN2B regulatory axis as a potential therapeutic target against human cardiac aging and aging-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yanxia Ye
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Kuan Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Haisong Liu
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Moshi Song
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Daoyuan Huang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yiyuan Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qun Chu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Yanling Fan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Sheng Zhang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yaobin Jing
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | - Si Wang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | | | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Weiqi Zhang
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China.
| | - Guang-Hui Liu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
33
|
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
|
34
|
Li Y, Ma Y, Gao L, Wang T, Zhuang Y, Zhang Y, Zheng L, Liu X. Upregulation of Microglial Sirt6 and Inhibition of Microglial Activation by Vitamin D3 in Lipopolysaccharide-stimulated Mice and BV-2 Cells. Neuroscience 2023; 526:85-96. [PMID: 37352968 DOI: 10.1016/j.neuroscience.2023.06.008] [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: 10/21/2022] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 06/25/2023]
Abstract
Vitamin D3 may suppress microglial activation and neuroinflammation, which play a central role in the pathophysiology of many neurological disorders. Sirt6 can remove histone 3 lysine 9 acetylation (H3K9ac) to repress expression of pathological genes and produce anti-inflammatory effects. However, whether vitamin D3 upregulates microglial Sirt6 to exert its protective effects against microglial activation and neuroinflammation is unclear. The effects of lower, normal, and higher dosages (1, 10 and 100 μg/kg/day) of vitamin D3 on behavioral and neuromorphological changes, brain inflammatory factors, Sirt6 and H3K9ac levels, and microglial Sirt6 distribution in hippocampus were evaluated in lipopolysaccharide (LPS)-stimulated mice. In addition, the effects of vitamin D3 on inflammatory factors, reactive oxygen species, Sirt6, and H3K9ac were confirmed in LPS-stimulated BV-2 cells. We verified that vitamin D3 ameliorated the impaired sociability of LPS-stimulated mice by three-chamber test. In addition, vitamin D3 upregulated brain Sirt6 generation, reduced H3K9ac levels and inhibited generation of brain inflammatory factors. Moreover, vitamin D3 promoted microglial Sirt6 distribution and attenuated microglia displaying an activated morphology in the hippocampus of LPS-stimulated mice. Similarly, vitamin D3 upregulated Sirt6 generation and intensity, reduced H3K9ac levels, and inhibited the inflammatory activation of LPS-stimulated BV-2 cells. In conclusion, vitamin D3 may upregulate microglial Sirt6 to reduce H3K9ac and inhibit microglial activation, thereby antagonizing neuroinflammation.
Collapse
Affiliation(s)
- Yanning Li
- Center for Drug Discovery Innovation, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei, People's Republic of China; Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, Hebei, People's Republic of China.
| | - Yujie Ma
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Lijie Gao
- Center for Drug Discovery Innovation, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei, People's Republic of China
| | - Ting Wang
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Yuchen Zhuang
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Yuping Zhang
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Long Zheng
- Department of Molecular Biology, Hebei Key Lab of Laboratory Animal Science, Hebei Medical University, Shijiazhuang 050017, Hebei, People's Republic of China
| | - Xifu Liu
- Center for Drug Discovery Innovation, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei, People's Republic of China.
| |
Collapse
|
35
|
Kirsch-Volders M, Fenech M. Towards prevention of aneuploidy-associated cellular senescence and aging: more questions than answers? MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2023; 792:108474. [PMID: 37866738 DOI: 10.1016/j.mrrev.2023.108474] [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: 02/15/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
The aim of this review is to discuss how aneuploidy contributes to the aging process, and to identify plausible strategies for its prevention. After an overview of mechanisms leading to aneuploidy and the major features of cellular senescence, we discuss the link between (i) aneuploidy and cellular senescence; (ii) aneuploidy and aging; and (iii) cellular senescence and aging. We also consider (i) interactions between aneuploidy, micronuclei, cellular senescence and aging, (ii) the potential of nutritional treatments to prevent aneuploidy-associated senescence and aging, and (iii) knowledge and technological gaps. Evidence for a causal link between aneuploidy, senescence and aging is emerging. In vitro, aneuploidy accompanies the entry into cellular senescence and can itself induce senescence. How aneuploidy contributes in vivo to cellular senescence is less clear. Several routes depending on aneuploidy and/or senescence converge towards chronic inflammation, the major driver of unhealthy aging. Aneuploidy can induce the pro-inflammatory Senescence Associated Secretory Phenotype (SASP), either directly or as a result of micronucleus (MN) induction leading to leakage of DNA into the cytoplasm and triggering of the cGAS-STING pathway of innate immune response. A major difficulty in understanding the impact of aneuploidy on senescence and aging in vivo, results from the heterogeneity of cellular senescence in different tissues at the cytological and molecular level. Due to this complexity, there is at the present time no biomarker or biomarker combination characteristic for all types of senescent cells. In conclusion, a deeper understanding of the critical role aneuploidy plays in cellular senescence and aging is essential to devise practical strategies to protect human populations from aneuploidy-associated pathologies. We discuss emerging evidence, based on in vitro and in vivo studies, that adequate amounts of specific micronutrients are essential for prevention of aneuploidy in humans and that precise nutritional intervention may be essential to help avoid the scourge of aneuploidy-driven diseases.
Collapse
Affiliation(s)
- Micheline Kirsch-Volders
- Laboratory for Cell Genetics, Department Biology, Faculty of Sciences and Bio-engineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Michael Fenech
- Clinical and Health Sciences, University of South Australia, SA 5000, Australia; Genome Health Foundation, North Brighton, SA 5048, Australia.
| |
Collapse
|
36
|
Zhang Y, Wang X, Li XK, Lv SJ, Wang HP, Liu Y, Zhou J, Gong H, Chen XF, Ren SC, Zhang H, Dai Y, Cai H, Yan B, Chen HZ, Tang X. Sirtuin 2 deficiency aggravates ageing-induced vascular remodelling in humans and mice. Eur Heart J 2023:ehad381. [PMID: 37377116 PMCID: PMC10393077 DOI: 10.1093/eurheartj/ehad381] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS The mechanisms underlying ageing-induced vascular remodelling remain unclear. This study investigates the role and underlying mechanisms of the cytoplasmic deacetylase sirtuin 2 (SIRT2) in ageing-induced vascular remodelling. METHODS AND RESULTS Transcriptome and quantitative real-time PCR data were used to analyse sirtuin expression. Young and old wild-type and Sirt2 knockout mice were used to explore vascular function and pathological remodelling. RNA-seq, histochemical staining, and biochemical assays were used to evaluate the effects of Sirt2 knockout on the vascular transcriptome and pathological remodelling and explore the underlying biochemical mechanisms. Among the sirtuins, SIRT2 had the highest levels in human and mouse aortas. Sirtuin 2 activity was reduced in aged aortas, and loss of SIRT2 accelerated vascular ageing. In old mice, SIRT2 deficiency aggravated ageing-induced arterial stiffness and constriction-relaxation dysfunction, accompanied by aortic remodelling (thickened vascular medial layers, breakage of elastin fibres, collagen deposition, and inflammation). Transcriptome and biochemical analyses revealed that the ageing-controlling protein p66Shc and metabolism of mitochondrial reactive oxygen species (mROS) contributed to SIRT2 function in vascular ageing. Sirtuin 2 repressed p66Shc activation and mROS production by deacetylating p66Shc at lysine 81. Elimination of reactive oxygen species by MnTBAP repressed the SIRT2 deficiency-mediated aggravation of vascular remodelling and dysfunction in angiotensin II-challenged and aged mice. The SIRT2 coexpression module in aortas was reduced with ageing across species and was a significant predictor of age-related aortic diseases in humans. CONCLUSION The deacetylase SIRT2 is a response to ageing that delays vascular ageing, and the cytoplasm-mitochondria axis (SIRT2-p66Shc-mROS) is important for vascular ageing. Therefore, SIRT2 may serve as a potential therapeutic target for vascular rejuvenation.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xiaoman Wang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xun-Kai Li
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Shuang-Jie Lv
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - He-Ping Wang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Yang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- Division of Vascular Surgery, Department of General Surgery, and Laboratory of Cardiovascular Diseases, West China Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| | - Jingyue Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| | - Hui Gong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| | - Xiao-Feng Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Si-Chong Ren
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, 783 Xindu Avenue, Chengdu, Sichuan 610500, China
| | - Huina Zhang
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Beijing 10029, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Yan
- Institute of Precision Medicine, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong 272067, China
| | - Hou-Zao Chen
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| |
Collapse
|
37
|
Lee MJ, Ryu HH, Hwang JW, Kim JR, Cho ES, Choi JK, Moon YJ. Sirt6 Activation Ameliorates Inflammatory Bone Loss in Ligature-Induced Periodontitis in Mice. Int J Mol Sci 2023; 24:10714. [PMID: 37445896 PMCID: PMC10341680 DOI: 10.3390/ijms241310714] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Periodontitis is an inflammatory disease caused by microorganisms that induce the destruction of periodontal tissue. Inflamed and damaged tissue produces various inflammatory cytokines, which activate osteoclasts and induce alveolar bone loss and, eventually, tooth loss. Sirt6 expression suppresses inflammation and bone resorption; however, its role in periodontitis remains unclear. We hypothesized that Sirt6 has a protective role in periodontitis. To understand the role of Sirt6 in periodontitis, we compared periodontitis with ligature placement around the maxillary left second molar in 8-week-old control (C57BL/6J) male mice to Sirt6-overexpressing Tg (Sirt6Tg) mice, and we observed the resulting phenotypes using micro-CT. MDL801, a Sirt6 activator, was used as a therapy for periodontitis through oral gavage. Pro-inflammatory cytokines and increased osteoclast numbers were observed in alveolar bone tissue under periodontitis surgery. In the same condition, interestingly, protein levels from Sirt6 were the most downregulated among sirtuins in alveolar bone tissue. Based on micro-CT and CEJ-ABC distance, Sirt6Tg was observed to resist bone loss against ligature-induced periodontitis. Furthermore, the number of osteoclasts was significantly reduced in Sirt6Tg-ligated mice compared with control-ligated mice, although systemic inflammatory cytokines did not change. Consistent with this observation, we confirmed that bone loss was significantly reduced when MDL801, a Sirt6 activator, was included in the ligation mouse model. Our findings demonstrate that Sirt6 activation prevents bone loss against ligature-induced periodontitis. Thus, a Sirt6 activator may provide a new therapeutic approach for periodontitis.
Collapse
Affiliation(s)
- Myung Jin Lee
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Hyang Hwa Ryu
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (H.H.R.); (J.W.H.)
| | - Jae Won Hwang
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (H.H.R.); (J.W.H.)
| | - Jung Ryul Kim
- Department of Orthopaedic Surgery, Jeonbuk National University Medical School and Hospital, Jeonju 54896, Republic of Korea;
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Jeonbuk National University School of Dentistry, Jeonju 54896, Republic of Korea;
| | - Jin Kyeong Choi
- Department of Immunology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Young Jae Moon
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea; (H.H.R.); (J.W.H.)
- Department of Orthopaedic Surgery, Jeonbuk National University Medical School and Hospital, Jeonju 54896, Republic of Korea;
| |
Collapse
|
38
|
Wang CL, Ohkubo R, Mu WC, Chen W, Fan JL, Song Z, Maruichi A, Sudmant PH, Pisco AO, Dubal DB, Ji N, Chen D. The mitochondrial unfolded protein response regulates hippocampal neural stem cell aging. Cell Metab 2023; 35:996-1008.e7. [PMID: 37146607 PMCID: PMC10330239 DOI: 10.1016/j.cmet.2023.04.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 04/14/2022] [Accepted: 04/06/2023] [Indexed: 05/07/2023]
Abstract
Aging results in a decline in neural stem cells (NSCs), neurogenesis, and cognitive function, and evidence is emerging to demonstrate disrupted adult neurogenesis in the hippocampus of patients with several neurodegenerative disorders. Here, single-cell RNA sequencing of the dentate gyrus of young and old mice shows that the mitochondrial protein folding stress is prominent in activated NSCs/neural progenitors (NPCs) among the neurogenic niche, and it increases with aging accompanying dysregulated cell cycle and mitochondrial activity in activated NSCs/NPCs in the dentate gyrus. Increasing mitochondrial protein folding stress results in compromised NSC maintenance and reduced neurogenesis in the dentate gyrus, neural hyperactivity, and impaired cognitive function. Reducing mitochondrial protein folding stress in the dentate gyrus of old mice improves neurogenesis and cognitive function. These results establish the mitochondrial protein folding stress as a driver of NSC aging and suggest approaches to improve aging-associated cognitive decline.
Collapse
Affiliation(s)
- Chih-Ling Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rika Ohkubo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wei-Chieh Mu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Wei Chen
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jiang Lan Fan
- Joint Graduate Program in Bioengineering, University of California, San Francisco, and University of California, Berkeley, San Francisco, CA 94720, USA
| | - Zehan Song
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ayane Maruichi
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Dena B Dubal
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Neurosciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Metabolic Biology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA; Endocrinology Graduate Program, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
39
|
Xiao F, Hu B, Si Z, Yang H, Xie J. Sirtuin 6 is a negative regulator of the anti-tumor function of natural killer cells in murine inflammatory colorectal cancer. Mol Immunol 2023; 158:68-78. [PMID: 37146480 DOI: 10.1016/j.molimm.2023.04.011] [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: 03/03/2023] [Revised: 04/20/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023]
Abstract
The immune system plays a crucial role in controlling colorectal cancer (CRC) development. Natural killer (NK) cells are tumoricidal but undergo exhaustion in CRC patients. The current research aims to understand the role of sirtuin 6 (SIRT6) in CRC-associated NK cell exhaustion in a murine inflammatory colorectal cancer model. To this end, inflammatory CRC was induced by treating mice with azoxymethane plus dextran sulfate sodium. The expression of SIRT6 in NK cells in murine mesenteric lymph nodes (mLNs) and the CRC tissue was characterized by Immunoblotting. SIRT6 knockdown was achieved by lentiviral transduction of murine splenic NK cells, followed by evaluation of NK cell proliferation and the expression of cytotoxic mediators using flow cytometry. NK cell cytotoxicity was measured by cytotoxicity assays. Adoptive transfer of murine NK cells was applied to analyze the effect of SIRT6 knockdown in vivo. We found that SIRT6 was up-regulated in infiltrating NK cells in the murine CRC tissue, especially NK cells with an exhausted phenotype and impaired cytotoxicity. SIRT6 knockdown significantly boosted murine splenic NK cell functionality, as evidenced by accelerated proliferation, increased production of cytotoxic mediators, and higher tumoricidal activity both in vitro and in vivo. Furthermore, the adoptive transfer of SIRT6-knockdown NK cells into CRC-bearing mice effectively suppressed CRC progression. Therefore, SIRT6 up-regulation is essential for murine NK cell exhaustion in CRC because it impedes the tumoricidal activity of murine NK cells. Artificial SIRT6 down-regulation could boost the function of infiltrating NK cells to oppress CRC progression in mice.
Collapse
Affiliation(s)
- Fei Xiao
- The Division of Gastrointestinal Surgery, Wuhan Fourth Hospital (Tongji Medical College Affiliated Wuhan Puai Hospital), 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei Province 430033, China
| | - Bo Hu
- The Division of Gastrointestinal Surgery, Wuhan Fourth Hospital (Tongji Medical College Affiliated Wuhan Puai Hospital), 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei Province 430033, China
| | - Zhilong Si
- The Division of Gastrointestinal Surgery, Wuhan Fourth Hospital (Tongji Medical College Affiliated Wuhan Puai Hospital), 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei Province 430033, China
| | - Huanbin Yang
- The Division of Gastrointestinal Surgery, Wuhan Fourth Hospital (Tongji Medical College Affiliated Wuhan Puai Hospital), 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei Province 430033, China
| | - Jun Xie
- The Division of Gastrointestinal Surgery, Wuhan Fourth Hospital (Tongji Medical College Affiliated Wuhan Puai Hospital), 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei Province 430033, China.
| |
Collapse
|
40
|
Sharma C, Donu D, Curry AM, Barton E, Cen Y. Multifunctional activity-based chemical probes for sirtuins. RSC Adv 2023; 13:11771-11781. [PMID: 37063743 PMCID: PMC10103746 DOI: 10.1039/d3ra02133e] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/18/2023] Open
Abstract
The sirtuin family of NAD+-dependent protein deacylases has gained significant attention during the last two decades, owing to their unique enzymatic activities as well as their critical roles in a broad array of cellular events. Innovative chemical probes are heavily pursued for the functional annotation and pharmacological perturbation of this group of "eraser" enzymes. We have developed several series of activity-based chemical probes (ABPs) to interrogate the functional state of active sirtuins in complex biological samples. They feature a simple Ala-Ala-Lys tripeptide backbone with a thioacyl "warhead", a photoaffinity group (benzophenone or diazirine), and a bioorthogonal group (terminal alkyne or azido) for conjugation to reporters. When applied in a comparative fashion, these probes reveal the changes of active sirtuin contents under different physiological conditions. Additionally, they can also be utilized in a competitive manner for inhibitor discovery. The Nobel-winning "click" conjugation to a fluorophore allows the visualization of the active enzymes, while the covalent adduct to a biotin leads to the affinity capture of the protein of interest. Furthermore, the "clickable" tag enables the easy access to proteolysis targeting chimeras (PROTACs) that effectively degrade human SIRT2 in HEK293 cells, albeit at micromolar concentrations. These small molecule probes offer unprecedented opportunities to investigate the biological functions and physiological relevance of the sirtuin family.
Collapse
Affiliation(s)
- Chiranjeev Sharma
- Department of Medicinal Chemistry, Virginia Commonwealth University Richmond VA 23219 USA +1-804-828-7405
| | - Dickson Donu
- Department of Medicinal Chemistry, Virginia Commonwealth University Richmond VA 23219 USA +1-804-828-7405
| | - Alyson M Curry
- Department of Medicinal Chemistry, Virginia Commonwealth University Richmond VA 23219 USA +1-804-828-7405
| | - Elizabeth Barton
- Department of Medicinal Chemistry, Virginia Commonwealth University Richmond VA 23219 USA +1-804-828-7405
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University Richmond VA 23219 USA +1-804-828-7405
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University Richmond VA 23219 USA
| |
Collapse
|
41
|
Yu J, Li T, Zhu J. Gene Therapy Strategies Targeting Aging-Related Diseases. Aging Dis 2023; 14:398-417. [PMID: 37008065 PMCID: PMC10017145 DOI: 10.14336/ad.2022.00725] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
Rapid advancements have taken place in gene therapy technology. However, effective methods for treating aging- or age-related chronic diseases, which are often closely related to genes or even multiple genes, are still lacking. The path to developing cures is winding, while gene therapy that targets genes related to aging represents an exciting research direction with tremendous potential. Among aging-related genes, some candidates have been studied at different levels, from cell to organismal levels (e.g., mammalian models) with different methods, from overexpression to gene editing. The TERT and APOE have even entered the stage of clinical trials. Even those displaying only a preliminary association with diseases have potential applications. This article discusses the foundations and recent breakthroughs in the field of gene therapy, providing a summary of current mainstream strategies and gene therapy products with clinical and preclinical applications. Finally, we review representative target genes and their potential for treating aging or age-related diseases.
Collapse
Affiliation(s)
| | | | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, National Key Laboratory for Medical Neurobiology, Institutes of Brain Science, Shanghai Key Laboratory of Brain Function and Regeneration, Institute of Neurosurgery, MOE Frontiers Center for Brain Science, Shanghai, China.
| |
Collapse
|
42
|
He Y, Su Y, Duan C, Wang S, He W, Zhang Y, An X, He M. Emerging role of aging in the progression of NAFLD to HCC. Ageing Res Rev 2023; 84:101833. [PMID: 36565959 DOI: 10.1016/j.arr.2022.101833] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 12/10/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
With the aging of global population, the incidence of nonalcoholic fatty liver disease (NAFLD) has surged in recent decades. NAFLD is a multifactorial disease that follows a progressive course, ranging from simple fatty liver, nonalcoholic steatohepatitis (NASH) to liver cirrhosis and hepatocellular carcinoma (HCC). It is well established that aging induces pathological changes in liver and potentiates the occurrence and progression of NAFLD, HCC and other age-related liver diseases. Studies of senescent cells also indicate a pivotal engagement in the development of NAFLD via diverse mechanisms. Moreover, nicotinamide adenine dinucleotide (NAD+), silence information regulator protein family (sirtuins), and mechanistic target of rapamycin (mTOR) are three vital and broadly studied targets involved in aging process and NAFLD. Nevertheless, the crucial role of these aging-associated factors in aging-related NAFLD remains underestimated. Here, we reviewed the current research on the roles of aging, cellular senescence and three aging-related factors in the evolution of NAFLD to HCC, aiming at inspiring promising therapeutic targets for aging-related NAFLD and its progression.
Collapse
Affiliation(s)
- Yongyuan He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinghong Su
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengcheng Duan
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyuan Wang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Basic Medicine, Kunming Medical University, China
| | - Yingting Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofei An
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Ming He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
43
|
Guo X, Su F, Gao Y, Tang L, Yu X, Zi J, Zhou Y, Wang H, Xue J, Wang X. Effects of dietary restriction on genome stability are sex and feeding regimen dependent. Food Funct 2023; 14:471-488. [PMID: 36519635 DOI: 10.1039/d2fo03138h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Preserving genome stability is essential to prevent aging and cancer. Dietary restriction (DR) is the most reproducible non-pharmacological way to improve health and extend lifespan in various species. Whether DR helps to preserve genome stability and whether this effect is altered by experimental variables remain unclear. Moreover, DR research relies heavily on experimental animals, making the development of reliable in vitro mimetics of great interest. Therefore, we tested the effects of sex and feeding regimen (time-restricted eating, alternate day fasting and calorie restriction) on genome stability in CF-1 mice and whether these effects can be recapitulated by cell culture paradigms. Here, we show that calorie restriction significantly decreases the spontaneous micronuclei (MN), a biomarker of genome instability, in bone marrow cells of females instead of males. Alternate day fasting significantly decreases cisplatin-induced MN in females instead of males. Unexpectedly, daily time-restricted eating significantly exacerbates cisplatin-induced MN in males but not in females. Additionally, we design several culture paradigms that are able to faithfully recapitulate the key effects of these DR regimens on genome stability. In particular, 30% reduction of serum, a mimetic of calorie restriction, exhibits a strong ability to decrease spontaneous and cisplatin-induced MN in immortalized human umbilical vein endothelial cells. We conclude that the effects of different DR regimens on genome stability are not universal and females from each diet regimen sustain a more stable genome than males. Our results provide novel insight into the understanding of how DR influences genome stability in a sex and regimen dependent way, and suggest that our in vitro DR mimetics could be adopted to study the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Xihan Guo
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China. .,Yunnan Environmental Mutagen Society, Kunming 650500, Yunnan, China
| | - Fuping Su
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China.
| | - Yue Gao
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China.
| | - Liyan Tang
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China.
| | - Xixi Yu
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China.
| | - Jiangli Zi
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China.
| | - Yingshui Zhou
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China.
| | - Han Wang
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China. .,Yunnan Environmental Mutagen Society, Kunming 650500, Yunnan, China
| | - Jinglun Xue
- Yeda Institute of Gene and Cell Therapy, Taizhou 318000, Zhejiang, China
| | - Xu Wang
- School of Life Sciences and The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming 650500, Yunnan, China. .,Yunnan Environmental Mutagen Society, Kunming 650500, Yunnan, China.,Yeda Institute of Gene and Cell Therapy, Taizhou 318000, Zhejiang, China
| |
Collapse
|
44
|
Zhu M, Yang X, Huang Y, Wang Z, Xiong Z. Serum SIRT6 Levels Are Associated with Frailty in Older Adults. J Nutr Health Aging 2023; 27:719-725. [PMID: 37754211 DOI: 10.1007/s12603-023-1969-y] [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: 03/15/2023] [Accepted: 07/23/2023] [Indexed: 09/28/2023]
Abstract
OBJECTIVES Frailty is one of the major health problems facing aging societies worldwide. We investigated the association between serum SIRT6 and frailty in older adults. DESIGN Cross-sectional analysis of associations of serum SIRT6 and frailty in older people. SETTING Enrolled community-dwelling and hospital outpatient clinic adults older than 65 years old in Wuhan City, Hubei Province, China. PARTICIPANTS A total of 540 community-dwelling older adults (age ≥ 65 years) in Wuhan were included in the study. MEASURES We used Frailty Phenotype criteria for classifying participants based on their frailty status. Serum SIRT6 was measured using an ELISA kit. RESULTS A total of 540 older adults were included in this cross-sectional study. Serum SIRT6 was lower in the slowness group (7.23±1.81 vs 5.89±1.74, p<0.001), weakness group (6.87±1.88 vs 5.68±1.64, p<0.001), and exhaustion group (6.73±1.90 vs 5.88±1.74, p<0.001) compare with the normal group. ROC curves were used to assess the efficiency of SIRT6 in predicting frailty in older adults. The AUC for SIRT6 was 0.792 (95% CI: 0.7514 to 0.8325), with the highest sensitivity of 68.0% and the specificity of 91.9%, and the optimal critical value of 4.65ng/ml according to Youden's index. Multivariate logistic regression analysis showed that serum SIRT6 level was independently associated with frailty in older people. CONCLUSION In conclusion, serum SIRT6 was decreased in frailty compared with robust older adults. A decreased serum SIRT6 was independently associated with an increased risk of frailty. SIRT6 may be a potential target for the treatment of patients with frailty.
Collapse
Affiliation(s)
- M Zhu
- Zhifan Xiong, Division of Gastroenterology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Science, Wuhan 430077, Hubei, China,
| | | | | | | | | |
Collapse
|
45
|
Shanmukha KD, Paluvai H, Lomada SK, Gokara M, Kalangi SK. Histone deacetylase (HDACs) inhibitors: Clinical applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:119-152. [DOI: 10.1016/bs.pmbts.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
46
|
Liu X, Ren S, Li Z, Hao D, Zhao X, Zhang Z, Liu D. Sirt6 mediates antioxidative functions by increasing Nrf2 abundance. Exp Cell Res 2023; 422:113409. [PMID: 36356655 DOI: 10.1016/j.yexcr.2022.113409] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/18/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Oxidative stress caused by excess ROS often leads to cellular macromolecule damage and eventually causes various biological catastrophes. Sirt6, a member of the mammalian homolog family of yeast Sir2 NAD+-dependent histone deacetylases, regulates multiple biological processes. Sirt6 exerts antioxidative functions by enhancing DNA repair and DNA end resection. In our study, we found that Sirt6 expression was induced by H2O2 and paraquat (PQ) in cells. When exposed to PQ, the Sirt6+/- C57BL/6 mice showed more serious liver damage and lower survival rate than the Sirt6+/+ mice. The Nrf2 protein levels and the mRNA levels of its target genes in mouse tissues were decreased by Sirt6 deficiency, and Sirt6 overexpression increased the Nrf2 protein content. Moreover, the endogenous H2O2 levels were increased in the tissues of Sirt6-deficient mice and were decreased in Sirt6 overexpression cells. Then, we found that Nrf2 was degraded faster in the Sirt6-deficient mouse embryonic fibroblasts (MEFs) than in the wild type MEFs and that Sirt6 enhanced the protein accumulation of Nrf2 in the nucleus. Lastly, we found that Sirt6 interacted with Nrf2 in co-IP and GST pull-down assays and that Sirt6 overexpression decreased the binding of Nrf2 to Keap1. Taken together, the results of the present study suggest that Sirt6 exerts antioxidative functions by increasing the Nrf2 protein level via Keap1-mediated regulation.
Collapse
Affiliation(s)
- Xiuzhen Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, PR China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256603, PR China
| | - Sichong Ren
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, PR China; Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, PR China
| | - Zuozhi Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, PR China
| | - Delong Hao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, PR China
| | - Xiang Zhao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, PR China
| | - Zhuqin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, PR China.
| | - Depei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, PR China.
| |
Collapse
|
47
|
Zeng J, Guo J, Huang S, Cheng Y, Luo F, Xu X, Chen R, Ma G, Wang Y. The roles of sirtuins in ferroptosis. Front Physiol 2023; 14:1131201. [PMID: 37153222 PMCID: PMC10157232 DOI: 10.3389/fphys.2023.1131201] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Ferroptosis represents a novel non-apoptotic form of regulated cell death that is driven by iron-dependent lipid peroxidation and plays vital roles in various diseases including cardiovascular diseases, neurodegenerative disorders and cancers. Plenty of iron metabolism-related proteins, regulators of lipid peroxidation, and oxidative stress-related molecules are engaged in ferroptosis and can regulate this complex biological process. Sirtuins have broad functional significance and are targets of many drugs in the clinic. Recently, a growing number of studies have revealed that sirtuins can participate in the occurrence of ferroptosis by affecting many aspects such as redox balance, iron metabolism, and lipid metabolism. This article reviewed the studies on the roles of sirtuins in ferroptosis and the related molecular mechanisms, highlighting valuable targets for the prevention and treatment of ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Jieqing Zeng
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Junhao Guo
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Si Huang
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Yisen Cheng
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Fei Luo
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Xusan Xu
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Riling Chen
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
| | - Guoda Ma
- Maternal and Children’s Health Research Institute, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
- *Correspondence: Guoda Ma, ; Yajun Wang,
| | - Yajun Wang
- Institute of Respiratory, Shunde Women and Children’s Hospital, Guangdong Medical University, Foshan, China
- *Correspondence: Guoda Ma, ; Yajun Wang,
| |
Collapse
|
48
|
Bhatiya M, Pathak S, Jothimani G, Duttaroy AK, Banerjee A. A Comprehensive Study on the Anti-cancer Effects of Quercetin and Its Epigenetic Modifications in Arresting Progression of Colon Cancer Cell Proliferation. Arch Immunol Ther Exp (Warsz) 2023; 71:6. [PMID: 36807774 PMCID: PMC9941246 DOI: 10.1007/s00005-023-00669-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/02/2022] [Indexed: 02/23/2023]
Abstract
Colon cancer etiology involves a wide spectrum of genetic and epigenetic alterations, finding it challenging to find effective therapeutic strategies. Quercetin exhibits potent anti-proliferative/apoptotic properties. In the present study, we aimed to elucidate the anti-cancer and anti-aging effect of quercetin in colon cancer cell lines. The anti-proliferative effect of quercetin was assessed in vitro by CCK-8 in normal and colon cancer cell lines. To check the anti-aging potential of quercetin, collagenase, elastase, and hyaluronidase inhibitory activity assays were performed. The epigenetic and DNA damage assays were performed using the human NAD-dependent deacetylase Sirtuin-6, proteasome 20S, Klotho, Cytochrome-C, and telomerase ELISA kits. Furthermore, the aging-associated miRNA expression profiling was performed on colon cancer cells. The treatment with quercetin inhibited cell proliferation of colon cancer cells in a dose-dependent manner. Quercetin arrested colon cancer cell growth by modulating expression of aging proteins including Sirtuin-6 and Klotho and also by inhibiting telomerase activity to restrict the telomere length which is evident from qPCR analysis. Quercetin also exhibited DNA damage protection by reducing proteasome 20S levels. The miRNA expression profiling results displayed differential expression of miRNA in colon cancer cell, and in addition, the highly upregulated miRNA was involved in the regulation of cell cycle, proliferation, and transcription. Our data suggest that quercetin treatment inhibited cell proliferation in colon cancer cells through regulating the anti-aging protein expression and provides better understanding for quercetin's potential use in colon cancer treatment.
Collapse
Affiliation(s)
- Meenu Bhatiya
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603 103 India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603 103 India
| | - Ganesan Jothimani
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603 103 India
| | - Asim K. Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu, 603 103, India.
| |
Collapse
|
49
|
Ziętara P, Dziewięcka M, Augustyniak M. Why Is Longevity Still a Scientific Mystery? Sirtuins-Past, Present and Future. Int J Mol Sci 2022; 24:728. [PMID: 36614171 PMCID: PMC9821238 DOI: 10.3390/ijms24010728] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
The sirtuin system consists of seven highly conserved regulatory enzymes responsible for metabolism, antioxidant protection, and cell cycle regulation. The great interest in sirtuins is associated with the potential impact on life extension. This article summarizes the latest research on the activity of sirtuins and their role in the aging process. The effects of compounds that modulate the activity of sirtuins were discussed, and in numerous studies, their effectiveness was demonstrated. Attention was paid to the role of a caloric restriction and the risks associated with the influence of careless sirtuin modulation on the organism. It has been shown that low modulators' bioavailability/retention time is a crucial problem for optimal regulation of the studied pathways. Therefore, a detailed understanding of the modulator structure and potential reactivity with sirtuins in silico studies should precede in vitro and in vivo experiments. The latest achievements in nanobiotechnology make it possible to create promising molecules, but many of them remain in the sphere of plans and concepts. It seems that solving the mystery of longevity will have to wait for new scientific discoveries.
Collapse
Affiliation(s)
| | | | - Maria Augustyniak
- Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, ul. Bankowa 9, 40-007 Katowice, Poland
| |
Collapse
|
50
|
Human centenarian-associated SIRT6 mutants modulate hepatocyte metabolism and collagen deposition in multilineage hepatic 3D spheroids. GeroScience 2022; 45:1177-1196. [PMID: 36534275 PMCID: PMC9886743 DOI: 10.1007/s11357-022-00713-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), encompassing fatty liver and its progression into nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC), is one of the rapidly rising health concerns worldwide. SIRT6 is an essential nuclear sirtuin that regulates numerous pathological processes including insulin resistance and inflammation, and recently it has been implicated in the amelioration of NAFLD progression. SIRT6 overexpression protects from formation of fibrotic lesions. However, the underlying molecular mechanisms are not fully delineated. Moreover, new allelic variants of SIRT6 (N308K/A313S) were recently associated with the longevity in Ashkenazi Jews by improving genome maintenance and DNA repair, suppressing transposons and killing cancer cells. Whether these new SIRT6 variants play different or enhanced roles in liver diseases is currently unknown. In this study, we aimed to clarify how these new centenarian-associated SIRT6 genetic variants affect liver metabolism and associated diseases. We present evidence that overexpression of centenarian-associated SIRT6 variants dramatically altered the metabolomic and secretomic profiles of unchallenged immortalized human hepatocytes (IHH). Most amino acids were increased in the SIRT6 N308K/A313S overexpressing IHH when compared to IHH transfected with the SIRT6 wild-type sequence. Several unsaturated fatty acids and glycerophospholipids were increased, and ceramide tended to be decreased upon SIRT6 N308K/A313S overexpression. Furthermore, we found that overexpression of SIRT6 N308K/A313S in a 3D hepatic spheroid model formed by the co-culture of human immortalized hepatocytes (IHH) and hepatic stellate cells (LX2) inhibited collagen deposition and fibrotic gene expression in absence of metabolic or dietary challenges. Hence, our findings suggest that novel longevity associated SIRT6 N308K/A313S variants could favor the prevention of NASH by altering hepatocyte proteome and lipidome.
Collapse
|