1
|
Jeayeng S, Thongsroy J, Chuaijit S. Caenorhabditis elegans as a Model to Study Aging and Photoaging. Biomolecules 2024; 14:1235. [PMID: 39456168 PMCID: PMC11505728 DOI: 10.3390/biom14101235] [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: 08/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Caenorhabditis elegans (C. elegans) has emerged as an outstanding model organism for investigating the aging process due to its shortened lifespan, well-defined genome, and accessibility of potent genetic tools. This review presents the current findings on chronological aging and photoaging in C. elegans, exploring the elaborate molecular pathways that control these processes. The progression of chronological aging is characterized by a gradual deterioration of physiological functions and is influenced by an interaction of genetic and environmental factors, including the insulin/insulin-like signaling (IIS) pathway. In contrast, photoaging is characterized by increased oxidative stress, DNA damage, and activation of stress response pathways induced by UV exposure. Although the genetic mechanisms of chronological aging in C. elegans have been characterized by extensive research, the pathways regulating photoaging are comparatively less well-studied. Here, we provide an overview of the current understanding of aging research, including the crucial genes and genetic pathways involved in the aging and photoaging processes of C. elegans. Understanding the complex interactions between these factors will provide invaluable insights into the molecular mechanisms underlying chronological aging and photoaging and may lead to novel therapeutic approaches and further studies for promoting healthy aging in humans.
Collapse
Affiliation(s)
- Saowanee Jeayeng
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat 80161, Thailand; (S.J.); (J.T.)
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80161, Thailand
| | - Jirapan Thongsroy
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat 80161, Thailand; (S.J.); (J.T.)
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80161, Thailand
| | - Sirithip Chuaijit
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat 80161, Thailand; (S.J.); (J.T.)
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80161, Thailand
| |
Collapse
|
2
|
Modarresi Chahardehi A, Afrooghe A, Emtiazi N, Rafiei S, Rezaei NJ, Dahmardeh S, Farz F, Naderi Z, Arefnezhad R, Motedayyen H. MicroRNAs and angiosarcoma: are there promising reports? Front Oncol 2024; 14:1385632. [PMID: 38826780 PMCID: PMC11143796 DOI: 10.3389/fonc.2024.1385632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
In recent years, microRNAs (miRNAs) have garnered increasing attention for their potential implications in cancer pathogenesis, functioning either as oncogenes or tumor suppressors. Notably, angiosarcoma, along with various other cardiovascular tumors such as lipomas, rhabdomyomas, hemangiomas, and myxomas, has shown variations in the expression of specific miRNA subtypes. A substantial body of evidence underscores the pivotal involvement of miRNAs in the genesis of angiosarcoma and certain cardiovascular tumors. This review aims to delve into the current literature on miRNAs and their prospective applications in cardiovascular malignancies, with a specific focus on angiosarcoma. It comprehensively covers diagnostic methods, prognostic evaluations, and potential treatments while providing a recapitulation of angiosarcoma's risk factors and molecular pathogenesis, with an emphasis on the role of miRNAs. These insights can serve as the groundwork for designing randomized control trials, ultimately facilitating the translation of these findings into clinical applications. Moving forward, it is imperative for studies to thoroughly scrutinize the advantages and disadvantages of miRNAs compared to current diagnostic and prognostic approaches in angiosarcoma and other cardiovascular tumors. Closing these knowledge gaps will be crucial for harnessing the full potential of miRNAs in the realm of angiosarcoma and cardiovascular tumor research.
Collapse
Affiliation(s)
| | - Arya Afrooghe
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikoo Emtiazi
- Department of Pathology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Rafiei
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | | | - Sarvin Dahmardeh
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Farz
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Naderi
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Arefnezhad
- Coenzyme R Research Institute, Tehran, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
3
|
Sharma G, Banerjee R, Srivastava S. Molecular Mechanisms and the Interplay of Important Chronic Obstructive Pulmonary Disease Biomarkers Reveals Novel Therapeutic Targets. ACS OMEGA 2023; 8:46376-46389. [PMID: 38107961 PMCID: PMC10719921 DOI: 10.1021/acsomega.3c07480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/02/2023] [Indexed: 12/19/2023]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a progressive, age-dependent, and unmet chronic inflammatory disease of the peripheral airways, leading to difficulty in exhalation. Several biomarkers have been tested in general towards the resolution for a long time, but no apparent success was achieved. Ongoing therapies of COPD have only symptomatic relief but no cure. Reactive oxygen species (ROS) are highly reactive species which include oxygen radicals and nonradical derivatives, and are the prominent players in COPD. They are produced as natural byproducts of cellular metabolism, but their levels can vary due to exposure to indoor air pollution, occupational pollution, and environmental pollutants such as cigarette smoke. In COPD, the lungs are continuously exposed to high levels of ROS thus leading to oxidative stress. ROS can cause damage to cells, proteins, lipids, and DNA which further contributes to the chronic inflammation in COPD and exacerbates the disease condition. Excessive ROS production can overwhelm cellular antioxidant systems and act as signaling molecules that regulate cellular processes, including antioxidant defense mechanisms involving glutathione and sirtuins which further leads to cellular apoptosis, cellular senescence, inflammation, and sarcopenia. In this review paper, we focused on COPD from different perspectives including potential markers and different cellular processes such as apoptosis, cellular senescence, inflammation, sirtuins, and sarcopenia, and tried to connect the dots between them so that novel therapeutic strategies to evaluate and target the possible underlying mechanisms in COPD could be explored.
Collapse
Affiliation(s)
- Gautam Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | | | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| |
Collapse
|
4
|
Copp ME, Shine J, Brown HL, Nimmala KR, Hansen OB, Chubinskaya S, Collins JA, Loeser RF, Diekman BO. Sirtuin 6 activation rescues the age-related decline in DNA damage repair in primary human chondrocytes. Aging (Albany NY) 2023; 15:13628-13645. [PMID: 38078876 PMCID: PMC10756124 DOI: 10.18632/aging.205394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
While advanced age is widely recognized as the greatest risk factor for osteoarthritis (OA), the biological mechanisms behind this connection remain unclear. Previous work has demonstrated that chondrocytes from older cadaveric donors have elevated levels of DNA damage as compared to chondrocytes from younger donors. The purpose of this study was to determine whether a decline in DNA repair efficiency is one explanation for the accumulation of DNA damage with age, and to quantify the improvement in repair with activation of Sirtuin 6 (SIRT6). After acute damage with irradiation, DNA repair was shown to be more efficient in chondrocytes from young (≤45 years old) as compared to middle-aged (50-65 years old) or older (>70 years old) cadaveric donors. Activation of SIRT6 with MDL-800 improved the repair efficiency, while inhibition with EX-527 reduced the rate of repair and increased the percentage of cells that retain high levels of damage. In addition to affecting repair after acute damage, treating chondrocytes from older donors with MDL-800 for 48 hours significantly reduced the amount of baseline DNA damage. Chondrocytes isolated from the knees of mice between 4 months and 22 months of age revealed both an increase in DNA damage with aging, and a decrease in DNA damage following MDL-800 treatment. Lastly, treating murine cartilage explants with MDL-800 lowered the percentage of chondrocytes with high p16 promoter activity, which supports the concept that using SIRT6 activation to maintain low levels of DNA damage may prevent the initiation of senescence.
Collapse
Affiliation(s)
- Michaela E. Copp
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hannon L. Brown
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Kirti R. Nimmala
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Oliver B. Hansen
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612, USA
| | - John A. Collins
- Department of Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA
| | - Richard F. Loeser
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian O. Diekman
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| |
Collapse
|
5
|
Chen R, Routh BN, Gaudet AD, Fonken LK. Circadian Regulation of the Neuroimmune Environment Across the Lifespan: From Brain Development to Aging. J Biol Rhythms 2023; 38:419-446. [PMID: 37357738 PMCID: PMC10475217 DOI: 10.1177/07487304231178950] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Circadian clocks confer 24-h periodicity to biological systems, to ultimately maximize energy efficiency and promote survival in a world with regular environmental light cycles. In mammals, circadian rhythms regulate myriad physiological functions, including the immune, endocrine, and central nervous systems. Within the central nervous system, specialized glial cells such as astrocytes and microglia survey and maintain the neuroimmune environment. The contributions of these neuroimmune cells to both homeostatic and pathogenic demands vary greatly across the day. Moreover, the function of these cells changes across the lifespan. In this review, we discuss circadian regulation of the neuroimmune environment across the lifespan, with a focus on microglia and astrocytes. Circadian rhythms emerge in early life concurrent with neuroimmune sculpting of brain circuits and wane late in life alongside increasing immunosenescence and neurodegeneration. Importantly, circadian dysregulation can alter immune function, which may contribute to susceptibility to neurodevelopmental and neurodegenerative diseases. In this review, we highlight circadian neuroimmune interactions across the lifespan and share evidence that circadian dysregulation within the neuroimmune system may be a critical component in human neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruizhuo Chen
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Brandy N. Routh
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Andrew D. Gaudet
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
- Department of Psychology, The University of Texas at Austin, Austin, Texas
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Laura K. Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
- Institute for Neuroscience, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
6
|
Copp ME, Shine J, Brown HL, Nimmala KR, Chubinskaya S, Collins JA, Loeser RF, Diekman BO. SIRT6 activation rescues the age-related decline in DNA damage repair in primary human chondrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530205. [PMID: 36909504 PMCID: PMC10002640 DOI: 10.1101/2023.02.27.530205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
While advanced age has long been recognized as the greatest risk factor for osteoarthritis (OA), the biological mechanisms behind this connection remain unclear. Previous work has demonstrated that chondrocytes from older cadaveric donors have elevated levels of DNA damage as compared to chondrocytes from younger donors. The purpose of this study was to determine whether a decline in DNA repair efficiency is one explanation for the accumulation of DNA damage with age, and to quantify the improvement in repair with activation of Sirtuin 6 (SIRT6). Using an acute irradiation model to bring the baseline level of all donors to the same starting point, this study demonstrates a decline in repair efficiency during aging when comparing chondrocytes from young (≤45 years old), middle-aged (50-65 years old), or older (>70 years old) cadaveric donors with no known history of OA or macroscopic cartilage degradation at isolation. Activation of SIRT6 in middle-aged chondrocytes with MDL-800 (20 μM) improved the repair efficiency, while inhibition with EX-527 (10 μM) inhibited the rate of repair and the increased the percentage of cells that retained high levels of damage. Treating chondrocytes from older donors with MDL-800 for 48 hours significantly reduced the amount of DNA damage, despite this damage having accumulated over decades. Lastly, chondrocytes isolated from the proximal femurs of mice between 4 months and 22 months of age revealed both an increase in DNA damage with aging, and a decrease in DNA damage following MDL-800 treatment.
Collapse
Affiliation(s)
- Michaela E Copp
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hannon L Brown
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| | - Kirti R Nimmala
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL
| | - John A Collins
- Department of Orthopedic Surgery, Thomas Jefferson University
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC
| |
Collapse
|
7
|
Dong W, Zhang K, Gong Z, Luo T, Li J, Wang X, Zou H, Song R, Zhu J, Ma Y, Liu G, Liu Z. N-acetylcysteine delayed cadmium-induced chronic kidney injury by activating the sirtuin 1-P53 signaling pathway. Chem Biol Interact 2023; 369:110299. [PMID: 36493885 DOI: 10.1016/j.cbi.2022.110299] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
With the development of modern industrial civilization, cadmium (Cd), a known nephrotoxic metal, has become a growing public safety issue due to its ability to induce various types of kidney disease. Maladaptive proximal tubule repair is a significant cause of Cd-induced chronic kidney disease (CKD), which is characterized by premature senescence and pro-fibrosis. Previously, we demonstrated that cadmium causes DNA damage and cycle arrest in renal tubular epithelial cells, which may be relevant to premature senescence regulated by sirtuin 1 (SIRT1). In this study, in vivo and in vitro studies were conducted to elucidate the role of SIRT1-mediated premature renal senescence in Cd-induced CKD. As oxidative stress is a significant cause of aging, we evaluated whether N-acetylcysteine (NAC) would inhibit Cd-induced premature aging and dysfunction in rat renal tubular epithelial cells. Cadmium induced premature renal senescence and fibrosis, and NAC inhibited premature renal senescence and fibrosis through the SIRT1-P53 pathway and delayed CKD progression. Overall, the results suggested that the SIRT1-P53 pathway mediates oxidative stress, premature renal senescence, and renal fibrosis during cadmium exposure, which may be a potential therapeutic target for Cd-induced CKD.
Collapse
Affiliation(s)
- Wenxuan Dong
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Kanglei Zhang
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Zhonggui Gong
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Tongwang Luo
- College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang Agriculture and Forestry University, Hangzhou, 311300, PR China
| | - Jiahui Li
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Xueru Wang
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Jiaqiao Zhu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China
| | - Gang Liu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China; Department of Pathology & Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, 12 East Wenhui Road, Yangzhou, 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, PR China.
| |
Collapse
|
8
|
Basova LV, Bortell N, Conti B, Fox HS, Milner R, Marcondes MCG. Age-associated changes in microglia activation and Sirtuin-1- chromatin binding patterns. Aging (Albany NY) 2022; 14:8205-8220. [PMID: 36227148 PMCID: PMC9648798 DOI: 10.18632/aging.204329] [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: 07/12/2022] [Accepted: 09/30/2022] [Indexed: 11/25/2022]
Abstract
The aging process is associated with changes in mechanisms maintaining physiology, influenced by genetics and lifestyle, and impacting late life quality and longevity. Brain health is critical in healthy aging. Sirtuin 1 (Sirt1), a histone deacetylase with silencing properties, is one of the molecular determinants experimentally linked to health and longevity. We compared brain pathogenesis and Sirt1-chromatin binding dynamics in brain pre-frontal cortex from 2 groups of elder rhesus macaques, divided by age of necropsy: shorter-lived animals (18-20 years old (yo)), equivalent to 60-70 human yo; and longer-lived animals (23-29 yo), corresponding to 80-100 human yo and modeling successful aging. These were compared with young adult brains (4-7 yo). Our findings indicated drastic differences in the microglia marker Iba1, along with factors influencing Sirt1 levels and activity, such as CD38 (an enzyme limiting NAD that controls Sirt1 activity) and mir142 (a microRNA targeting Sirt1 transcription) between the elder groups. Iba1 was lower in shorter-lived animals than in the other groups, while CD38 was higher in both aging groups compared to young. mir142 and Sirt1 levels were inversely correlated in longer-lived brains (>23yo), but not in shorter-lived brains (18-20 yo). We also found that Sirt1 binding showed signs of better efficiency in longer-lived animals compared to shorter-lived ones, in genes associated with nuclear activity and senescence. Overall, differences in neuroinflammation and Sirt1 interactions with chromatin distinguished shorter- and longer-lived animals, suggesting the importance of preserving microglia and Sirt1 functional efficiency for longevity.
Collapse
Affiliation(s)
- Liana V. Basova
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
| | | | - Bruno Conti
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
| | - Howard S. Fox
- University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Richard Milner
- San Diego Biomedical Research Institute, San Diego, CA 92121, USA
| | | |
Collapse
|
9
|
Cummings MJ, Yu H, Paudel S, Hu G, Li X, Hemberger M, Wang X. Uterine-specific SIRT1 deficiency confers premature uterine aging and impairs invasion and spacing of blastocyst, and stromal cell decidualization, in mice. Mol Hum Reprod 2022; 28:gaac016. [PMID: 35536234 PMCID: PMC10689003 DOI: 10.1093/molehr/gaac016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/28/2022] [Indexed: 12/02/2023] Open
Abstract
A distinct age-related alteration in the uterine environment has recently been identified as a prevalent cause of the reproductive decline in older female mice. However, the molecular mechanisms that underlie age-associated uterine adaptability to pregnancy are not known. Sirtuin 1 (SIRT1), a multifunctional NAD+-dependent deacetylase that regulates cell viability, senescence and inflammation during aging, is reduced in aged decidua. Thus, we hypothesize that SIRT1 plays a critical role in uterine adaptability to pregnancy and that uterine-specific ablation of Sirt1 gene accelerates premature uterine aging. Female mice with uterine ablation of Sirt1 gene using progesterone receptor Cre (PgrCre) exhibit subfertility and signs of premature uterine aging. These Sirt1-deficient mothers showed decreases in litter size from their 1st pregnancy and became sterile (25.1 ± 2.5 weeks of age) after giving birth to the third litter. We report that uterine-specific Sirt1 deficiency impairs invasion and spacing of blastocysts, and stromal cell decidualization, leading to abnormal placentation. We found that these problems traced back to the very early stages of hormonal priming of the uterus. During the window of receptivity, Sirt1 deficiency compromises uterine epithelial-stromal crosstalk, whereby estrogen, progesterone and Indian hedgehog signaling pathways are dysregulated, hampering stromal cell priming for decidualization. Uterine transcriptomic analyses also link these causes to perturbations of histone proteins and epigenetic modifiers, as well as adrenomedullin signaling, hyaluronic acid metabolism, and cell senescence. Strikingly, our results also identified genes with significant overlaps with the transcriptome of uteri from aged mice and transcriptomes related to master regulators of decidualization (e.g. Foxo1, Wnt4, Sox17, Bmp2, Egfr and Nr2f2). Our results also implicate accelerated deposition of aging-related fibrillar Type I and III collagens in Sirt1-deficient uteri. Collectively, SIRT1 is an important age-related regulator of invasion and spacing of blastocysts, as well as decidualization of stromal cells.
Collapse
Affiliation(s)
- Magdalina J Cummings
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Hongyao Yu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sudikshya Paudel
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Myriam Hemberger
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Xiaoqiu Wang
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
10
|
Ungurianu A, Zanfirescu A, Margină D. Regulation of Gene Expression through Food—Curcumin as a Sirtuin Activity Modulator. PLANTS 2022; 11:plants11131741. [PMID: 35807694 PMCID: PMC9269530 DOI: 10.3390/plants11131741] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 12/24/2022]
Abstract
The sirtuin family comprises NAD+-dependent protein lysine deacylases, mammalian sirtuins being either nuclear (SIRT1, SIRT2, SIRT6, and SIRT7), mitochondrial (SIRT3, SIRT4, and SIRT5) or cytosolic enzymes (SIRT2 and SIRT5). They are able to catalyze direct metabolic reactions, thus regulating several physiological functions, such as energy metabolism, stress response, inflammation, cell survival, DNA repair, tissue regeneration, neuronal signaling, and even circadian rhythms. Based on these data, recent research was focused on finding molecules that could regulate sirtuins’ expression and/or activity, natural compounds being among the most promising in the field. Curcumin (1,7-bis-(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) can induce, through SIRT, modulation of cancer cell senescence, improve endothelial cells protection against atherosclerotic factors, enhance muscle regeneration in atrophy models, and act as a pro-longevity factor counteracting the neurotoxicity of amyloid-beta. Although a plethora of protective effects was reported (antioxidant, anti-inflammatory, anticancer, etc.), its therapeutical use is limited due to its bioavailability issues. However, all the reported effects may be explained via the bioactivation theory, which postulates that curcumin’s observed actions are modulated via its metabolites and/or degradation products. The present article is focused on bringing together the literature data correlating the ability of curcumin and its metabolites to modulate SIRT activity and its consequent beneficial effects.
Collapse
Affiliation(s)
- Anca Ungurianu
- Department of Biochemistry, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Traian Vuia, 020956 Bucharest, Romania; (A.U.); (D.M.)
| | - Anca Zanfirescu
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Traian Vuia, 020956 Bucharest, Romania
- Correspondence:
| | - Denisa Margină
- Department of Biochemistry, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Traian Vuia, 020956 Bucharest, Romania; (A.U.); (D.M.)
| |
Collapse
|
11
|
Modeling of the Senescence-Associated Phenotype in Human Skin Fibroblasts. Int J Mol Sci 2022; 23:ijms23137124. [PMID: 35806127 PMCID: PMC9266450 DOI: 10.3390/ijms23137124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/28/2022] Open
Abstract
Modern understanding of aging is based on the accumulation of cellular damage during one’s life span due to the gradual deterioration of regenerative mechanisms in response to the continuous effect of stress, lifestyle, and environmental factors, followed by increased morbidity and mortality. Simultaneously, the number of senescent cells accumulate exponentially as organisms age. Cell culture models are valuable tools to investigate the mechanisms of aging by inducing cellular senescence in stress-induced premature senescence (SIPS) models. Here, we explain the three-step and one-step H2O2-induced senescence models of SIPS designed and reproduced on different human dermal fibroblast cell lines (CCD-1064Sk, CCD-1135Sk, and BJ-5ta). In both SIPS models, it was evident that the fibroblasts developed similar aging characteristics as cells with replicative senescence. Among the most noticeable senescent biomarkers were increased β-Gal expression, high levels of the p21 protein, altered levels of cell-cycle regulators (i.e., CDK2 and c-Jun), compromised extracellular matrix (ECM) composition, reduced cellular viability, and delayed wound healing properties. Based on the significant increase in senescence biomarkers in fibroblast cultures, reduced functional activity, and metabolic dysfunction, the one-step senescence model was chosen as a feasible and reliable method for future testing of anti-aging compounds.
Collapse
|
12
|
Pukhalskaia AE, Diatlova AS, Linkova NS, Kvetnoy IM. Sirtuins: Role in the Regulation of Oxidative Stress and the Pathogenesis of Neurodegenerative Diseases. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2022; 52:164-174. [DOI: 10.1007/s11055-022-01217-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/23/2020] [Accepted: 09/28/2020] [Indexed: 01/03/2025]
|
13
|
Gonzales MM, Krishnamurthy S, Garbarino V, Daeihagh AS, Gillispie GJ, Deep G, Craft S, Orr ME. A geroscience motivated approach to treat Alzheimer's disease: Senolytics move to clinical trials. Mech Ageing Dev 2021; 200:111589. [PMID: 34687726 PMCID: PMC9059898 DOI: 10.1016/j.mad.2021.111589] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 12/29/2022]
Abstract
The pathogenic processes driving Alzheimer's disease (AD) are complex. An incomplete understanding of underlying disease mechanisms has presented insurmountable obstacles for developing effective disease-modifying therapies. Advanced chronological age is the greatest risk factor for developing AD. Intervening on biological aging may alter disease progression and represents a novel, complementary approach to current strategies. Toward this end, cellular senescence has emerged as a promising target. This complex stress response harbors damaged cells in a cell cycle arrested, apoptosis-resistant cell state. Senescent cells accumulate with age where they notoriously secrete molecules that contribute to chronic tissue dysfunction and disease. Thus, benefits of cell survival in a senescent fate are countered by their toxic secretome. The removal of senescent cells improves brain structure and function in rodent models at risk of developing AD, and in those with advanced Aβ and tau pathology. The present review describes the path to translating this promising treatment strategy to AD clinical trials. We review evidence for senescent cell accumulation in the human brain, considerations and strategies for senescence-targeting trials specific to AD, approaches to detect senescent brain cells in biofluids, and summarize the goals of the first senolytic trials for the treatment of AD (NCT04063124 and NCT04685590). This article is part of the Special Issue - Senolytics - Edited by Joao Passos and Diana Jurk.
Collapse
Affiliation(s)
- Mitzi M Gonzales
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sudarshan Krishnamurthy
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Bowman Gray Center for Medical Education, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Valentina Garbarino
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ali S Daeihagh
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Gregory J Gillispie
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Miranda E Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA; Salisbury VA Medical Center, Salisbury, NC, USA.
| |
Collapse
|
14
|
Gonzales MM, Krishnamurthy S, Garbarino V, Daeihagh AS, Gillispie GJ, Deep G, Craft S, Orr ME. A geroscience motivated approach to treat Alzheimer’s disease: Senolytics move to clinical trials. Mech Ageing Dev 2021. [DOI: 10.1016/j.mad.2021.111589
expr 868687188 + 807217478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
15
|
Junker N, Gossmann TI. Adaptation-Driven Evolution of Sirtuin 1 (SIRT1), a Key Regulator of Metabolism and Aging, in Marmot Species. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.666564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The sirtuin protein family plays a role in the lifespan of various species and is involved in numerous key metabolic processes. To understand the evolutionary role of sirtuins in marmots, a long-living rodent species group with remarkable metabolic shutdown during hibernation, we conducted a phylogeny-based substitution rate analysis of coding genes based on genetic information of seven marmot species. We show that sirtuin 1 (SIRT1) has evolved under positive selection in the marmot lineage. We pinpoint three amino acid changes in four different marmot species that underlie the signal of positive selection and that may favor increased longevity in marmots. Based on a computational structural analysis we can show that all three substitutions affect the secondary structure of the same region in human SIRT1. We propose that the identified region is close to the catalytic domain and that the potential structural changes may impact the catalytic activity of the enzyme and therefore might be playing a functional role in marmot's extended lifespan and metabolic shutdown.
Collapse
|
16
|
Wang L, Zuo X, Ouyang Z, Qiao P, Wang F. A Systematic Review of Antiaging Effects of 23 Traditional Chinese Medicines. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:5591573. [PMID: 34055012 PMCID: PMC8143881 DOI: 10.1155/2021/5591573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Aging is an inevitable stage of body development. At the same time, aging is a major cause of cancer, cardiovascular disease, and neurodegenerative diseases. Chinese herbal medicine is a natural substance that can effectively delay aging and is expected to be developed as antiaging drugs in the future. Aim of the review. This paper reviews the antiaging effects of 23 traditional Chinese herbal medicines or their active components. Materials and methods. We reviewed the literature published in the last five years on Chinese herbal medicines or their active ingredients and their antiaging role obtained through the following databases: PubMed, EMBASE, Scopus, and Web of Science. RESULTS A total of 2485 papers were found, and 212 papers were screened after removing the duplicates and reading the titles. Twenty-three studies met the requirements of this review and were included. Among these studies, 13 articles used Caenorhabditis elegans as the animal model, and 10 articles used other animal models or cell lines. CONCLUSION Chinese herbal medicines or their active components play an antiaging role by regulating genes related to aging through a variety of signaling pathways. Chinese herbal medicines are expected to be developed as antiaging drugs or used in the medical cosmetology industry.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xu Zuo
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhuoer Ouyang
- Department of Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ping Qiao
- Department of Cell Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
17
|
Di Daniele N, Marrone G, Di Lauro M, Di Daniele F, Palazzetti D, Guerriero C, Noce A. Effects of Caloric Restriction Diet on Arterial Hypertension and Endothelial Dysfunction. Nutrients 2021; 13:nu13010274. [PMID: 33477912 PMCID: PMC7833363 DOI: 10.3390/nu13010274] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
The most common manifestation of cardiovascular (CV) diseases is the presence of arterial hypertension (AH), which impacts on endothelial dysfunction. CV risk is associated with high values of systolic and diastolic blood pressure and depends on the presence of risk factors, both modifiable and not modifiable, such as overweight, obesity, physical exercise, smoking, age, family history, and gender. The main target organs affected by AH are the heart, brain, vessels, kidneys, and eye retina. AH onset can be counteracted or delayed by adopting a proper diet, characterized by a low saturated fat and sodium intake, a high fruit and vegetable intake, a moderate alcohol consumption, and achieving and maintaining over time the ideal body weight. In this review, we analyzed how a new nutritional approach, named caloric restriction diet (CRD), can provide a significant reduction in blood pressure values and an improvement of the endothelial dysfunction. In fact, CRD is able to counteract aging and delay the onset of CV and neurodegenerative diseases through the reduction of body fat mass, systolic and diastolic values, free radicals production, and oxidative stress. Currently, there are few studies on CRD effects in the long term, and it would be advisable to perform observational studies with longer follow-up.
Collapse
Affiliation(s)
- Nicola Di Daniele
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (F.D.D.); (D.P.); (C.G.); (A.N.)
- Correspondence: ; Tel.: +39-062090-2982; Fax: +39-062090-3362
| | - Giulia Marrone
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (F.D.D.); (D.P.); (C.G.); (A.N.)
- School of Applied Medical, Surgical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Manuela Di Lauro
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (F.D.D.); (D.P.); (C.G.); (A.N.)
| | - Francesca Di Daniele
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (F.D.D.); (D.P.); (C.G.); (A.N.)
- School of Applied Medical, Surgical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Daniela Palazzetti
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (F.D.D.); (D.P.); (C.G.); (A.N.)
| | - Cristina Guerriero
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (F.D.D.); (D.P.); (C.G.); (A.N.)
| | - Annalisa Noce
- UOC of Internal Medicine-Center of Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.M.); (M.D.L.); (F.D.D.); (D.P.); (C.G.); (A.N.)
| |
Collapse
|
18
|
Sims CA, Labiner HE, Shah SS, Baur JA. Longevity pathways in stress resistance: targeting NAD and sirtuins to treat the pathophysiology of hemorrhagic shock. GeroScience 2021; 43:1217-1228. [PMID: 33462707 DOI: 10.1007/s11357-020-00311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/06/2020] [Indexed: 11/29/2022] Open
Abstract
Stress resistance correlates with longevity and this pattern has been exploited to help identify genes that can influence lifespan. Reciprocally, genes and pharmacological agents that have been studied primarily in the context of longevity may be an untapped resource for treating acute stresses. Here we summarize the evidence that targeting SIRT1, studied primarily in the context of longevity, can improve outcomes in hemorrhagic shock and resuscitation. Hemorrhagic shock is a potentially fatal condition that occurs when blood loss is so severe that tissues no longer receive adequate oxygen. While stabilizing the blood pressure and reperfusing tissues are necessary, re-introducing oxygen to ischemic tissues generates a burst of reactive oxygen species that can cause secondary tissue damage. Reactive oxygen species not only exacerbate the inflammatory cascade but also can directly damage mitochondria, leading to bioenergetic failure in the affected tissues. Treatments with polyphenol resveratrol and with nicotinamide adenine dinucleotide (NAD) precursors have both shown promising results in rodent models of hemorrhagic shock and resuscitation. Although a number of different mechanisms may be at play in each case, a common theme is that resveratrol and NAD both enhance the activity of SIRT1. Moreover, many of the physiologic improvements observed with resveratrol and NAD precursors are consistent with modulation of known SIRT1 targets. Because small blood vessels and limited blood volume make mice very challenging for the development of hemorrhagic shock models, there is a paucity of direct genetic evidence testing the role of SIRT1. However, the development of more robust methods in mice as well as genetic modifications in rats should allow the study of SIRT1 transgenic and KO rodents in the near future. The potential therapeutic effect of SIRT1 in hemorrhagic shock may serve as an important example supporting the value of considering "longevity" pathways in the mitigation of acute stresses.
Collapse
Affiliation(s)
- Carrie A Sims
- Division of Trauma, Critical Care, and Burn at The Ohio State University Wexner Medical Center, The Ohio State University, Faculty Office Tower, 395 12th Ave, Room 654, Columbus, OH, 43210, USA.
| | - Hanna E Labiner
- Division of Trauma, Critical Care, and Burn at The Ohio State University Wexner Medical Center, The Ohio State University, Faculty Office Tower, 395 12th Ave, Room 654, Columbus, OH, 43210, USA
| | - Sohini S Shah
- Institute for Diabetes, Obesity and Metabolism and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., SCTR 12-114, Philadelphia, PA, 19104, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity and Metabolism and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., SCTR 12-114, Philadelphia, PA, 19104, USA.
| |
Collapse
|
19
|
Meng J, Liu Y, Xie Z, Qing H, Lei P, Ni J. Nucleus distribution of cathepsin B in senescent microglia promotes brain aging through degradation of sirtuins. Neurobiol Aging 2020; 96:255-266. [PMID: 33049518 DOI: 10.1016/j.neurobiolaging.2020.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/10/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
Abstract
Cathepsin B (CatB) leakage from the lysosome into the cytosol in senescent microglia is associated with cognitive impairment. However, whether cellular compartmental translocation of CatB is associated with brain aging remains unclear. In the present study, increased CatB was found in the nucleus of CatB-overexpressed microglia followed by L-leucyl-L-leucine methyl ester, a lysosome-destabilizing reagent, and in the nuclear fraction of the cortex and hippocampus from aged mice. Moreover, CatB enzymatic activity examination showed the nuclear CatB exhibited the proteolytic activity to cleave its specific substrates. The amount of sirtuin1 (Sirt1), Sirt6, Sirt7, and p-Sirt1 was decreased in the cortical lysates from aged mice, in parallel with increased expression of proinflammatory mediators, which were diminished by CatB deficiency. Furthermore, intralateral ventricle administration of microglia overexpressed CatB, and treatment with L-leucyl-L-leucine methyl ester induced cognitive impairment in middle-aged mice. These observations suggest that the increase and nucleus translocation of CatB in senescent microglia were involved in the degradation of nuclear Sirts, which induced proinflammatory responses, resulting in cognition impairment.
Collapse
Affiliation(s)
- Jie Meng
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yicong Liu
- The Affiliated Stomatology Hospital, School of Medical, Zhejiang University, Zhejiang, China
| | - Zhen Xie
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China.
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China; Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
20
|
Zhao L, Cao J, Hu K, He X, Yun D, Tong T, Han L. Sirtuins and their Biological Relevance in Aging and Age-Related Diseases. Aging Dis 2020; 11:927-945. [PMID: 32765955 PMCID: PMC7390530 DOI: 10.14336/ad.2019.0820] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/20/2019] [Indexed: 12/18/2022] Open
Abstract
Sirtuins, initially described as histone deacetylases and gene silencers in yeast, are now known to have many more functions and to be much more abundant in living organisms. The increasing evidence of sirtuins in the field of ageing and age-related diseases indicates that they may provide novel targets for treating diseases associated with aging and perhaps extend human lifespan. Here, we summarize some of the recent discoveries in sirtuin biology that clearly implicate the functions of sirtuins in the regulation of aging and age-related diseases. Furthermore, human sirtuins are considered promising therapeutic targets for anti-aging and ageing-related diseases and have attracted interest in scientific communities to develop small molecule activators or drugs to ameliorate a wide range of ageing disorders. In this review, we also summarize the discovery and development status of sirtuin-targeted drug and further discuss the potential medical strategies of sirtuins in delaying aging and treating age-related diseases.
Collapse
Affiliation(s)
- Lijun Zhao
- 1Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, China
| | - Jianzhong Cao
- 2Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kexin Hu
- 1Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, China
| | - Xiaodong He
- 2Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dou Yun
- 1Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, China
| | - Tanjun Tong
- 1Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, China
| | - Limin Han
- 1Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, China
| |
Collapse
|
21
|
Albert E, Laimins L. Regulation of the Human Papillomavirus Life Cycle by DNA Damage Repair Pathways and Epigenetic Factors. Viruses 2020; 12:E744. [PMID: 32664381 PMCID: PMC7412114 DOI: 10.3390/v12070744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023] Open
Abstract
Human papillomaviruses are the causative agents of cervical and other anogenital cancers along with approximately 60% of oropharyngeal cancers. These small double-stranded DNA viruses infect stratified epithelia and link their productive life cycles to differentiation. HPV proteins target cellular factors, such as those involved in DNA damage repair, as well as epigenetic control of host and viral transcription to regulate the productive life cycle. HPVs constitutively activate the ATM and ATR DNA repair pathways and preferentially recruit these proteins to viral genomes to facilitate productive viral replication. In addition, the sirtuin deacetylases along with histone acetyltransferases, including Tip60, are targeted in HPV infections to regulate viral transcription and replication. These pathways provide potential targets for drug therapy to treat HPV-induced disease.
Collapse
Affiliation(s)
| | - Laimonis Laimins
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA;
| |
Collapse
|
22
|
Mir SM, Samavarchi Tehrani S, Goodarzi G, Jamalpoor Z, Asadi J, Khelghati N, Qujeq D, Maniati M. Shelterin Complex at Telomeres: Implications in Ageing. Clin Interv Aging 2020; 15:827-839. [PMID: 32581523 PMCID: PMC7276337 DOI: 10.2147/cia.s256425] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/18/2020] [Indexed: 12/16/2022] Open
Abstract
Different factors influence the development and control of ageing. It is well known that progressive telomere shorting is one of the molecular mechanisms underlying ageing. The shelterin complex consists of six telomere-specific proteins which are involved in the protection of chromosome ends. More particularly, this vital complex protects the telomeres from degradation, prevents from activation of unwanted repair systems, regulates the activity of telomerase, and has a crucial role in cellular senescent and ageing-related pathologies. This review explores the organization and function of telomeric DNA along with the mechanism of telomeres during ageing, followed by a discussion of the critical role of shelterin components and their changes during ageing.
Collapse
Affiliation(s)
- Seyed Mostafa Mir
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran.,Student Research Committee, Babol University of Medical Sciences, Babol, Iran.,Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Jamalpoor
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Jahanbakhsh Asadi
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Nafiseh Khelghati
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Durdi Qujeq
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran.,Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mahmood Maniati
- School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
23
|
Sirtuins family as a target in endothelial cell dysfunction: implications for vascular ageing. Biogerontology 2020; 21:495-516. [PMID: 32285331 DOI: 10.1007/s10522-020-09873-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/20/2020] [Indexed: 12/13/2022]
Abstract
The vascular endothelium is a protective barrier between the bloodstream and the vasculature that may be disrupted by different factors such as the presence of diseased states. Diseases like diabetes and obesity pose a great risk toward endothelial cell inflammation and oxidative stress, leading to endothelial cell dysfunction and thereby cardiovascular complications such as atherosclerosis. Sirtuins are NAD+-dependent histone deacetylases that are implicated in the pathophysiology of cardiovascular diseases, and they have been identified to be important regulators of endothelial cell function. A handful of recent studies suggest that disbalance in the regulation of endothelial sirtuins, mainly sirtuin 1 (SIRT1), contributes to endothelial cell dysfunction. Herein, we summarize how SIRT1 and other sirtuins may contribute to endothelial cell function and how presence of diseased conditions may alter their expressions to cause endothelial dysfunction. Moreover, we discuss how the beneficial effects of exercise on the endothelium are dependent on SIRT1. These mainly include regulation of signaling pathways related to endothelial nitric oxide synthase phosphorylation and nitric oxide production, mitochondrial biogenesis and mitochondria-mediated apoptotic pathways, oxidative stress and inflammatory pathways. Sirtuins as modulators of the adverse conditions in the endothelium hold a promising therapeutic potential for health conditions related to endothelial dysfunction and vascular ageing.
Collapse
|
24
|
Sirtuins and SIRT6 in Carcinogenesis and in Diet. Int J Mol Sci 2019; 20:ijms20194945. [PMID: 31591350 PMCID: PMC6801518 DOI: 10.3390/ijms20194945] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Sirtuins are a highly conserved family of nicotinamide adenine dinucleotide (NAD)-dependent protein lysine modifying enzymes. They are key regulators for a wide variety of cellular and physiological processes such as cell proliferation, differentiation, DNA damage and stress response, genome stability, cell survival, metabolism, energy homeostasis, organ development and aging. Aging is one of the major risk factors of cancer, as many of the physiological mechanisms and pathologies associated with the aging process also contribute to tumor initiation, growth and/or metastasis. This review focuses on one the mammalian sirtuins, SIRT6, which has emerged as an important regulator of longevity and appears to have multiple biochemical functions that interfere with tumor development and may be useful in cancer prevention and for site-specific treatment. The recent evidence of the role of SIRT6 in carcinogenesis is also discussed, focusing on the potential use of SIRT6 modulators in cancer nanomedicine.
Collapse
|
25
|
SIRT1 Modulators in Experimentally Induced Liver Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8765954. [PMID: 31281594 PMCID: PMC6589266 DOI: 10.1155/2019/8765954] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/21/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
Abstract
This article is directed at highlighting the involvement of the endogenous stress sensor SIRT1 (silent information regulator T1) as a possible factor involved in hepatoprotection. The selective SIRT1 modulators whether activators (STACs) or inhibitors are being tried experimentally and clinically. We discuss the modulation of SIRT1 on cytoprotection or even cytotoxicity in the liver chemically injured by hepatotoxic agents in rats, to shed light on the crosstalk between SIRT1 and its modulators. A combination of D-galactosamine and lipopolysaccharide (D-GalN/LPS) downregulated SIRT1 expression, while SIRT1 activators, SRT1720, resveratrol, and quercetin, upregulated SIRT1 and alleviated D-GalN/LPS-induced acute hepatotoxicity. Liver injury markers exhibited an inverse relationship with SIRT1 expression. However, under subchronic hepatotoxicity, quercetin decreased the significant increase in SIRT1 expression to lower levels which are still higher than normal ones and mitigated the liver-damaging effects of carbon tetrachloride. Each of these STACs was hepatoprotective and returned the conventional antioxidant enzymes to the baseline. Polyphenols tend to fine-tune SIRT1 expression towards normal in the liver of intoxicated rats in both acute and subchronic studies. Together, all these events give an impression that the cytoprotective effects of SIRT1 are exhibited within a definite range of expression. The catalytic activity of SIRT1 is important in the hepatoprotective effects of polyphenols where SIRT1 inhibitors block and the allosteric SIRT1 activators mimic the hepatoprotective effects of polyphenols. Our findings indicate that the pharmacologic modulation of SIRT1 could represent both an important move in alleviating hepatic insults and a future major step in the treatment of xenobiotic-induced hepatotoxicity.
Collapse
|
26
|
Abstract
Sirtuin is an essential factor that delays cellular senescence and extends the organismal lifespan through the regulation of diverse cellular processes. Suppression of cellular senescence by Sirtuin is mainly mediated through delaying the age-related telomere attrition, sustaining genome integrity and promotion of DNA damage repair. In addition, Sirtuin modulates the organismal lifespan by interacting with several lifespan regulating signaling pathways including insulin/IGF-1 signaling pathway, AMP-activated protein kinase, and forkhead box O. Although still controversial, it is suggested that the prolongevity effect of Sirtuin is dependent with the level of and with the tissue expression of Sirtuin. Since Sirtuin is also believed to mediate the prolongevity effect of calorie restriction, activators of Sirtuin have attracted the attention of researchers to develop therapeutics for age-related diseases. Resveratrol, a phytochemical rich in the skin of red grapes and wine, has been actively investigated to activate Sirtuin activity with consequent beneficial effects on aging. This article reviews the evidences and controversies regarding the roles of Sirtuin on cellular senescence and lifespan extension, and summarizes the activators of Sirtuin including Sirtuin-activating compounds and compounds that increase the cellular level of nicotinamide dinucleotide.
Collapse
Affiliation(s)
- Shin-Hae Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Ji-Hyeon Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Hye-Yeon Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Kyung-Jin Min
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| |
Collapse
|
27
|
Sirtuin1 Targeting Reverses Innate and Adaptive Immune Tolerance in Septic Mice. J Immunol Res 2018; 2018:2402593. [PMID: 30069485 PMCID: PMC6057336 DOI: 10.1155/2018/2402593] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/29/2018] [Indexed: 01/08/2023] Open
Abstract
Resistance and tolerance to infection are two universal fitness and survival strategies used by inflammation and immunity in organisms and cells to guard homeostasis. During sepsis, however, both strategies fail, and animal and human victims often die from combined innate and adaptive immune suppression with persistent bacterial and viral infections. NAD+-sensing nuclear sirtuin1 (SIRT1) epigenetically guards immune and metabolic homeostasis during sepsis. Pharmacologically inhibiting SIRT1 deacetylase activity in septic mice reverses monocyte immune tolerance, clears infection, rebalances glycolysis and glucose oxidation, resolves organ dysfunction, and prevents most septic deaths. Whether SIRT1 inhibition during sepsis treatment concomitantly reverses innate and T cell antigen-specific immune tolerance is unknown. Here, we show that treating septic mice with a SIRT1 selective inhibitor concordantly reverses immune tolerance splenic dendritic and antigen-specific tolerance of splenic CD4+ and CD8+ T cells. SIRT1 inhibition also increases the ratio of IL12 p40+ and TNFα proinflammatory/immune to IL10 and TGFβ anti-inflammatory/immune cytokines and decreases the ratio of CD4+ TReg repressor to CD4+ activator T cells. These findings support the unifying concept that nuclear NAD+ sensor SIRT1 broadly coordinates innate and adaptive immune reprogramming during sepsis and is a druggable immunometabolic enhancement target.
Collapse
|
28
|
Paredes S, Angulo-Ibanez M, Tasselli L, Carlson SM, Zheng W, Li TM, Chua KF. The epigenetic regulator SIRT7 guards against mammalian cellular senescence induced by ribosomal DNA instability. J Biol Chem 2018; 293:11242-11250. [PMID: 29728458 DOI: 10.1074/jbc.ac118.003325] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/21/2018] [Indexed: 12/16/2022] Open
Abstract
In the yeast Saccharomyces cerevisiae, genomic instability in rDNA repeat sequences is an underlying cause of cell aging and is suppressed by the chromatin-silencing factor Sir2. In humans, rDNA instability is observed in cancers and premature aging syndromes, but its underlying mechanisms and functional consequences remain unclear. Here, we uncovered a pivotal role of sirtuin 7 (SIRT7), a mammalian Sir2 homolog, in guarding against rDNA instability and show that this function of SIRT7 protects against senescence in primary human cells. We found that, mechanistically, SIRT7 is required for association of SNF2H (also called SMARCA5, SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily A, member 5), a component of the nucleolar heterochromatin-silencing complex NoRC, with rDNA sequences. Defective rDNA-heterochromatin silencing in SIRT7-deficient cells unleashed rDNA instability, with excision and loss of rDNA gene copies, which in turn induced acute senescence. Mounting evidence indicates that accumulation of senescent cells significantly contributes to tissue dysfunction in aging-related pathologies. Our findings identify rDNA instability as a driver of mammalian cellular senescence and implicate SIRT7-dependent heterochromatin silencing in protecting against this process.
Collapse
Affiliation(s)
- Silvana Paredes
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Maria Angulo-Ibanez
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Luisa Tasselli
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Scott M Carlson
- Department of Biology, Stanford University, Stanford, California 94305
| | - Wei Zheng
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Tie-Mei Li
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305.,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| | - Katrin F Chua
- From the Department of Medicine, Stanford University School of Medicine, Stanford, California 94305, .,Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, and
| |
Collapse
|
29
|
Dai H, Sinclair DA, Ellis JL, Steegborn C. Sirtuin activators and inhibitors: Promises, achievements, and challenges. Pharmacol Ther 2018; 188:140-154. [PMID: 29577959 DOI: 10.1016/j.pharmthera.2018.03.004] [Citation(s) in RCA: 328] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The NAD+-dependent protein lysine deacylases of the Sirtuin family regulate various physiological functions, from energy metabolism to stress responses. The human Sirtuin isoforms, SIRT1-7, are considered attractive therapeutic targets for aging-related diseases, such as type 2 diabetes, inflammatory diseases and neurodegenerative disorders. We review the status of Sirtuin-targeted drug discovery and development. Potent and selective pharmacological Sirt1 activators and inhibitors are available, and initial clinical trials have been carried out. Several promising inhibitors and activators have also been described for other isoforms. Progress in understanding the mechanisms of Sirtuin modulation by such compounds provides a rational basis for further drug development.
Collapse
Affiliation(s)
- Han Dai
- GlaxoSmithKline, 1250S. Collegeville Road, Collegeville, PA 19426, USA
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School, Boston, MA 02115, USA
| | - James L Ellis
- GlaxoSmithKline, 1250S. Collegeville Road, Collegeville, PA 19426, USA
| | - Clemens Steegborn
- Department of Biochemistry and Research Center for Bio-Macromolecules, University of Bayreuth, 95440 Bayreuth, Germany.
| |
Collapse
|
30
|
Zhang Q, Zhang P, Qi GJ, Zhang Z, He F, Lv ZX, Peng X, Cai HW, Li TX, Wang XM, Tian B. Cdk5 suppression blocks SIRT1 degradation via the ubiquitin-proteasome pathway in Parkinson's disease models. Biochim Biophys Acta Gen Subj 2018; 1862:1443-1451. [PMID: 29571747 DOI: 10.1016/j.bbagen.2018.03.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/18/2022]
Abstract
The NAD+-dependent protein deacetylase sirtuin 1 (SIRT1), a member of the sirtuin family, may have a neuroprotective effect in multiple neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS). Many studies have suggested that overexpression-induced or resveratrol-treated activation of SIRT1 could significantly ameliorate several neurodegenerative diseases in mouse models. However, the type of SIRT1, protein expression levels and underlying mechanisms remain unclear, especially in PD. In this study, the results demonstrated that SIRT1 knockout markedly worsened the movement function in MPTP-lesioned animal model of PD. SIRT1 expression was found to be markedly decreased not only in environmental factor PD models, neurotoxin MPP+-treated primary culture neurons and MPTP-induced mice but also in genetic factor PD models, overexpressed α-synuclein-A30PA53T SH-SY5Y stable cell line and hm2α-SYN-39 transgenic mouse strain. Importantly, the degradation of SIRT1 during MPP+ treatment was mediated by the ubiquitin-proteasome pathway. Furthermore, the results indicated that cyclin-dependent kinase 5 (Cdk5) was also involved in the decrease of SIRT1 expression, which could be efficiently blocked by the inhibition of Cdk5. In conclusion, our findings revealed that the Cdk5-dependent ubiquitin-proteasome pathway mediated degradation of SIRT1 plays a vital role in the progression of PD.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Pei Zhang
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Guang-Jian Qi
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Zheng Zhang
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Feng He
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Ze-Xi Lv
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Xiang Peng
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Hong-Wei Cai
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Tong-Xia Li
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China
| | - Xue-Min Wang
- Department of Neurobiology, Southern Medical University, Guangzhou, Guangdong Province 510515, PR China
| | - Bo Tian
- Department of Neurobiology, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China; Key Laboratory of Neurological Diseases, Ministry of Education, 13 Hangkong Road, Wuhan, Hubei Province 430030, PR China; Institute for Brain Research, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, PR China.
| |
Collapse
|
31
|
Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid Redox Signal 2018; 28:643-661. [PMID: 28891317 PMCID: PMC5824489 DOI: 10.1089/ars.2017.7290] [Citation(s) in RCA: 519] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Antioxidant and redox signaling (ARS) events are regulated by critical molecules that modulate antioxidants, reactive oxygen species (ROS) or reactive nitrogen species (RNS), and/or oxidative stress within the cell. Imbalances in these molecules can disturb cellular functions to become pathogenic. Sirtuins serve as important regulators of ARS in cells. Recent Advances: Sirtuins (SIRTs 1-7) are a family of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylases with the ability to deacetylate histone and nonhistone targets. Recent studies show that sirtuins modulate the regulation of a variety of cellular processes associated with ARS. SIRT1, SIRT3, and SIRT5 protect the cell from ROS, and SIRT2, SIRT6, and SIRT7 modulate key oxidative stress genes and mechanisms. Interestingly, SIRT4 has been shown to induce ROS production and has antioxidative roles as well. CRITICAL ISSUES A complete understanding of the roles of sirtuins in redox homeostasis of the cell is very important to understand the normal functioning as well as pathological manifestations. In this review, we have provided a critical discussion on the role of sirtuins in the regulation of ARS. We have also discussed mechanistic interactions among different sirtuins. Indeed, a complete understanding of sirtuin biology could be critical at multiple fronts. FUTURE DIRECTIONS Sirtuins are emerging to be important in normal mammalian physiology and in a variety of oxidative stress-mediated pathological situations. Studies are needed to dissect the mechanisms of sirtuins in maintaining redox homeostasis. Efforts are also required to assess the targetability of sirtuins in the management of redox-regulated diseases. Antioxid. Redox Signal. 28, 643-661.
Collapse
Affiliation(s)
- Chandra K Singh
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | | | - Nicholas J Mack
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| |
Collapse
|
32
|
Sims CA, Baur JA. The grapes and wrath: using resveratrol to treat the pathophysiology of hemorrhagic shock. Ann N Y Acad Sci 2017; 1403:70-81. [PMID: 28845517 DOI: 10.1111/nyas.13444] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/28/2017] [Accepted: 07/03/2017] [Indexed: 12/17/2022]
Abstract
Resveratrol, a naturally occurring polyphenol found in grapes, has been shown to reduce oxidative stress and inflammation in a variety of conditions. Recently, resveratrol has been investigated as a potential adjunct to resuscitation therapy for hemorrhagic shock-a condition characterized by tissue hypoxia, mitochondrial dysfunction, and inflammation. Although standard resuscitation restores tissue perfusion, it can exacerbate oxidative stress and organ damage. In rodent models of severe hemorrhagic shock, resveratrol mitigates reperfusion injury, preserves organ function, and improves survival. While many of these benefits can be attributed to its ability to activate sirtuin 1, resveratrol interacts with many targets that are relevant to ischemia-reperfusion. Here, we explore the probable mechanisms, potential benefits, and possible problems associated with administering resveratrol as an adjunct during resuscitation of hemorrhagic shock.
Collapse
Affiliation(s)
- Carrie A Sims
- The Trauma Center at the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph A Baur
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
33
|
Bonkowski MS, Sinclair DA. Slowing ageing by design: the rise of NAD + and sirtuin-activating compounds. Nat Rev Mol Cell Biol 2016; 17:679-690. [PMID: 27552971 DOI: 10.1038/nrm.2016.93] [Citation(s) in RCA: 564] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The sirtuins (SIRT1-7) are a family of nicotinamide adenine dinucleotide (NAD+)-dependent deacylases with remarkable abilities to prevent diseases and even reverse aspects of ageing. Mice engineered to express additional copies of SIRT1 or SIRT6, or treated with sirtuin-activating compounds (STACs) such as resveratrol and SRT2104 or with NAD+ precursors, have improved organ function, physical endurance, disease resistance and longevity. Trials in non-human primates and in humans have indicated that STACs may be safe and effective in treating inflammatory and metabolic disorders, among others. These advances have demonstrated that it is possible to rationally design molecules that can alleviate multiple diseases and possibly extend lifespan in humans.
Collapse
Affiliation(s)
- Michael S Bonkowski
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - David A Sinclair
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Pharmacology, The University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
34
|
Salminen A, Kaarniranta K, Hiltunen M, Kauppinen A. Krebs cycle dysfunction shapes epigenetic landscape of chromatin: Novel insights into mitochondrial regulation of aging process. Cell Signal 2014; 26:1598-603. [DOI: 10.1016/j.cellsig.2014.03.030] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/30/2014] [Indexed: 02/09/2023]
|
35
|
Miteva YV, Cristea IM. A proteomic perspective of Sirtuin 6 (SIRT6) phosphorylation and interactions and their dependence on its catalytic activity. Mol Cell Proteomics 2013; 13:168-83. [PMID: 24163442 DOI: 10.1074/mcp.m113.032847] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Sirtuin 6 (SIRT6), a member of the mammalian sirtuin family, is a nuclear deacetylase with substrate-specific NAD(+)-dependent activity. SIRT6 has emerged as a critical regulator of diverse processes, including DNA repair, gene expression, telomere maintenance, and metabolism. However, our knowledge regarding its interactions and regulation remains limited. Here, we present a comprehensive proteomics-based analysis of SIRT6 protein interactions and their dependence on SIRT6 catalytic activity. We also identify evolutionarily conserved SIRT6 phosphorylations, including four within a proline-rich disordered region, and show that the conserved S338 phosphorylation can modulate selected SIRT6 interactions. By integrating molecular biology tools, microscopy, immunoaffinity purifications, label-free quantitative mass spectrometry, and bioinformatic analyses, we have established the first large-scale SIRT6 interaction network. Relative protein abundances and gene ontology functional assessment highlighted proteins involved in transcription regulation, chromatin organization, nuclear transport, telomerase function, and RNA processing. Independent immunoisolations under increased stringency distinguished the most stable SIRT6 interactions. One prominent interaction with Ras-GTPase-activating protein-binding protein 1 (G3BP1) was further validated by microscopy, reciprocal purifications, and isolations in different cell types and of endogenous SIRT6. Interestingly, a subset of specific interactions, including G3BP1, were significantly reduced or abolished in isolations of catalytically deficient SIRT6 mutant, revealing previously unknown interplay between SIRT6 activity and its associations. Overall, our study reveals putative means of regulation of SIRT6 functions via interactions and modifications, providing an important resource for future studies on the molecular mechanisms underlying sirtuin functions.
Collapse
Affiliation(s)
- Yana V Miteva
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | | |
Collapse
|
36
|
Abstract
The mammalian sirtuins (SIRT1-7) are NAD(+)-dependent lysine deacylases that play central roles in cell survival, inflammation, energy metabolism, and aging. Members of this family of enzymes are considered promising pharmaceutical targets for the treatment of age-related diseases including cancer, type 2 diabetes, inflammatory disorders, and Alzheimer's disease. SIRT1-activating compounds (STACs), which have been identified from a variety of chemical classes, provide health benefits in animal disease models. Recent data point to a common mechanism of allosteric activation by natural and synthetic STACs that involves the binding of STACs to a conserved N-terminal domain in SIRT1. Compared with polyphenols such as resveratrol, the synthetic STACs show greater potency, solubility, and target selectivity. Although considerable progress has been made regarding SIRT1 allosteric activation, key questions remain, including how the molecular contacts facilitate SIRT1 activation, whether other sirtuin family members will be amenable to activation, and whether STACs will ultimately prove safe and efficacious in humans.
Collapse
Affiliation(s)
- David A Sinclair
- Glenn Laboratories for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115;
| | | |
Collapse
|
37
|
SIRT6 recruits SNF2H to DNA break sites, preventing genomic instability through chromatin remodeling. Mol Cell 2013; 51:454-68. [PMID: 23911928 DOI: 10.1016/j.molcel.2013.06.018] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 05/16/2013] [Accepted: 06/24/2013] [Indexed: 11/20/2022]
Abstract
DNA damage is linked to multiple human diseases, such as cancer, neurodegeneration, and aging. Little is known about the role of chromatin accessibility in DNA repair. Here, we find that the deacetylase sirtuin 6 (SIRT6) is one of the earliest factors recruited to double-strand breaks (DSBs). SIRT6 recruits the chromatin remodeler SNF2H to DSBs and focally deacetylates histone H3K56. Lack of SIRT6 and SNF2H impairs chromatin remodeling, increasing sensitivity to genotoxic damage and recruitment of downstream factors such as 53BP1 and breast cancer 1 (BRCA1). Remarkably, SIRT6-deficient mice exhibit lower levels of chromatin-associated SNF2H in specific tissues, a phenotype accompanied by DNA damage. We demonstrate that SIRT6 is critical for recruitment of a chromatin remodeler as an early step in the DNA damage response, indicating that proper unfolding of chromatin plays a rate-limiting role. We present a unique crosstalk between a histone modifier and a chromatin remodeler, regulating a coordinated response to prevent DNA damage.
Collapse
|
38
|
Cosentino C, Mostoslavsky R. Metabolism, longevity and epigenetics. Cell Mol Life Sci 2013; 70:1525-41. [PMID: 23467663 PMCID: PMC3625512 DOI: 10.1007/s00018-013-1295-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 02/05/2013] [Accepted: 02/05/2013] [Indexed: 12/21/2022]
Abstract
Metabolic homeostasis and interventions that influence nutrient uptake are well-established means to influence lifespan even in higher eukaryotes. Until recently, the molecular mechanisms explaining such an effect remained scantily understood. Sirtuins are a group of protein deacetylases that depend on the metabolic intermediate NAD(+) as a cofactor for their function. For this reason they sense metabolic stress and in turn function at multiple levels to exert proper metabolic adaptation. Among other things, sirtuins can perform as histone deacetylases inducing epigenetic changes to modulate transcription and DNA repair. Recent studies have indicated that beyond sirtuins, the activity of other chromatin modifiers, such as histone acetyl transferases, might also be tightly linked to the availability of their intermediate metabolite acetyl-CoA. We summarize current knowledge of the emerging concepts indicating close crosstalk between the epigenetic machineries able to sense metabolic stress, their adaptive metabolic responses and their potential role in longevity.
Collapse
Affiliation(s)
- Claudia Cosentino
- The Massachusetts General Hospital Cancer Center-Harvard Medical School, 185 Cambridge St, Boston, MA USA
| | - Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center-Harvard Medical School, 185 Cambridge St, Boston, MA USA
| |
Collapse
|
39
|
White AT, Schenk S. NAD(+)/NADH and skeletal muscle mitochondrial adaptations to exercise. Am J Physiol Endocrinol Metab 2012; 303:E308-21. [PMID: 22436696 PMCID: PMC3423123 DOI: 10.1152/ajpendo.00054.2012] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 03/15/2012] [Indexed: 12/21/2022]
Abstract
The pyridine nucleotides, NAD(+) and NADH, are coenzymes that provide oxidoreductive power for the generation of ATP by mitochondria. In skeletal muscle, exercise perturbs the levels of NAD(+), NADH, and consequently, the NAD(+)/NADH ratio, and initial research in this area focused on the contribution of redox control to ATP production. More recently, numerous signaling pathways that are sensitive to perturbations in NAD(+)(H) have come to the fore, as has an appreciation for the potential importance of compartmentation of NAD(+)(H) metabolism and its subsequent effects on various signaling pathways. These pathways, which include the sirtuin (SIRT) proteins SIRT1 and SIRT3, the poly(ADP-ribose) polymerase (PARP) proteins PARP1 and PARP2, and COOH-terminal binding protein (CtBP), are of particular interest because they potentially link changes in cellular redox state to both immediate, metabolic-related changes and transcriptional adaptations to exercise. In this review, we discuss what is known, and not known, about the contribution of NAD(+)(H) metabolism and these aforementioned proteins to mitochondrial adaptations to acute and chronic endurance exercise.
Collapse
Affiliation(s)
- Amanda T White
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
40
|
White AT, Schenk S. NAD(+)/NADH and skeletal muscle mitochondrial adaptations to exercise. AMERICAN JOURNAL OF PHYSIOLOGY. ENDOCRINOLOGY AND METABOLISM 2012. [PMID: 22436696 DOI: 10.1152/ajpendo.00054.2012.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pyridine nucleotides, NAD(+) and NADH, are coenzymes that provide oxidoreductive power for the generation of ATP by mitochondria. In skeletal muscle, exercise perturbs the levels of NAD(+), NADH, and consequently, the NAD(+)/NADH ratio, and initial research in this area focused on the contribution of redox control to ATP production. More recently, numerous signaling pathways that are sensitive to perturbations in NAD(+)(H) have come to the fore, as has an appreciation for the potential importance of compartmentation of NAD(+)(H) metabolism and its subsequent effects on various signaling pathways. These pathways, which include the sirtuin (SIRT) proteins SIRT1 and SIRT3, the poly(ADP-ribose) polymerase (PARP) proteins PARP1 and PARP2, and COOH-terminal binding protein (CtBP), are of particular interest because they potentially link changes in cellular redox state to both immediate, metabolic-related changes and transcriptional adaptations to exercise. In this review, we discuss what is known, and not known, about the contribution of NAD(+)(H) metabolism and these aforementioned proteins to mitochondrial adaptations to acute and chronic endurance exercise.
Collapse
Affiliation(s)
- Amanda T White
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
41
|
SIRT1 negatively regulates the activities, functions, and protein levels of hMOF and TIP60. Mol Cell Biol 2012; 32:2823-36. [PMID: 22586264 DOI: 10.1128/mcb.00496-12] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
SIRT1 is a NAD(+)-dependent histone H4K16 deacetylase that controls several different normal physiologic and disease processes. Like most histone deacetylases, SIRT1 also deacetylates nonhistone proteins. Here, we show that two members of the MYST (MOZ, Ybf2/Sas3, Sas2, and TIP60) acetyltransferase family, hMOF and TIP60, are SIRT1 substrates. SIRT1 deacetylation of the enzymatic domains of hMOF and TIP60 inhibits their acetyltransferase activity and promotes ubiquitination-dependent degradation of these proteins. Importantly, immediately following DNA damage, the binding of SIRT1 to hMOF and TIP60 is transiently interrupted, with corresponding hMOF/TIP60 hyperacetylation. Lysine-to-arginine mutations in SIRT1-targeted lysines on hMOF and TIP60 repress DNA double-strand break repair and inhibit the ability of hMOF/TIP60 to induce apoptosis in response to DNA double-strand break. Together, these findings uncover novel pathways in which SIRT1 dynamically interacts with and regulates hMOF and TIP60 through deacetylation and provide additional mechanistic insights by which SIRT1 regulates DNA damage response.
Collapse
|