1
|
Engin A. Misalignment of Circadian Rhythms in Diet-Induced Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:27-71. [PMID: 39287848 DOI: 10.1007/978-3-031-63657-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronize them with daily cycles. While the central clock in the suprachiasmatic nucleus (SCN) is mainly synchronized by the light/dark cycles, the peripheral clocks react to other stimuli, including the feeding/fasting state, nutrients, sleep-wake cycles, and physical activity. During the disruption of circadian rhythms due to genetic mutations or social and occupational obligations, incorrect arrangement between the internal clock system and environmental rhythms leads to the development of obesity. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition leads to uncoupling of the peripheral clocks from the central pacemaker and to the development of metabolic disorders. The strong coupling of the SCN to the light-dark cycle creates a situation of misalignment when food is ingested during the "wrong" time of day. Food-anticipatory activity is mediated by a self-sustained circadian timing, and its principal component is a food-entrainable oscillator. Modifying the time of feeding alone greatly affects body weight, whereas ketogenic diet (KD) influences circadian biology, through the modulation of clock gene expression. Night-eating behavior is one of the causes of circadian disruption, and night eaters have compulsive and uncontrolled eating with severe obesity. By contrast, time-restricted eating (TRE) restores circadian rhythms through maintaining an appropriate daily rhythm of the eating-fasting cycle. The hypothalamus has a crucial role in the regulation of energy balance rather than food intake. While circadian locomotor output cycles kaput (CLOCK) expression levels increase with high-fat diet-induced obesity, peroxisome proliferator-activated receptor-alpha (PPARα) increases the transcriptional level of brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like 1 (BMAL1) in obese subjects. In this context, effective timing of chronotherapies aiming to correct SCN-driven rhythms depends on an accurate assessment of the SCN phase. In fact, in a multi-oscillator system, local rhythmicity and its disruption reflects the disruption of either local clocks or central clocks, thus imposing rhythmicity on those local tissues, whereas misalignment of peripheral oscillators is due to exosome-based intercellular communication.Consequently, disruption of clock genes results in dyslipidemia, insulin resistance, and obesity, while light exposure during the daytime, food intake during the daytime, and sleeping during the biological night promote circadian alignment between the central and peripheral clocks. Thus, shift work is associated with an increased risk of obesity, diabetes, and cardiovascular diseases because of unusual eating times as well as unusual light exposure and disruption of the circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
2
|
Bhasin S, Seals D, Migaud M, Musi N, Baur JA. Nicotinamide Adenine Dinucleotide in Aging Biology: Potential Applications and Many Unknowns. Endocr Rev 2023; 44:1047-1073. [PMID: 37364580 DOI: 10.1210/endrev/bnad019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/28/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Recent research has unveiled an expansive role of NAD+ in cellular energy generation, redox reactions, and as a substrate or cosubstrate in signaling pathways that regulate health span and aging. This review provides a critical appraisal of the clinical pharmacology and the preclinical and clinical evidence for therapeutic effects of NAD+ precursors for age-related conditions, with a particular focus on cardiometabolic disorders, and discusses gaps in current knowledge. NAD+ levels decrease throughout life; age-related decline in NAD+ bioavailability has been postulated to be a contributor to many age-related diseases. Raising NAD+ levels in model organisms by administration of NAD+ precursors improves glucose and lipid metabolism; attenuates diet-induced weight gain, diabetes, diabetic kidney disease, and hepatic steatosis; reduces endothelial dysfunction; protects heart from ischemic injury; improves left ventricular function in models of heart failure; attenuates cerebrovascular and neurodegenerative disorders; and increases health span. Early human studies show that NAD+ levels can be raised safely in blood and some tissues by oral NAD+ precursors and suggest benefit in preventing nonmelanotic skin cancer, modestly reducing blood pressure and improving lipid profile in older adults with obesity or overweight; preventing kidney injury in at-risk patients; and suppressing inflammation in Parkinson disease and SARS-CoV-2 infection. Clinical pharmacology, metabolism, and therapeutic mechanisms of NAD+ precursors remain incompletely understood. We suggest that these early findings provide the rationale for adequately powered randomized trials to evaluate the efficacy of NAD+ augmentation as a therapeutic strategy to prevent and treat metabolic disorders and age-related conditions.
Collapse
Affiliation(s)
- Shalender Bhasin
- Department of Medicine, Harvard Medical School, Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Douglas Seals
- Department of Integrative Physiology and Medicine, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Marie Migaud
- Department of Pharmacology, Mitchell Cancer Institute, College of Medicine, University of Southern Alabama, Mobile, AL 36688, USA
| | - Nicolas Musi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joseph A Baur
- Department of Physiology, Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Xu Y, Xiao W. NAD+: An Old but Promising Therapeutic Agent for Skeletal Muscle Ageing. Ageing Res Rev 2023; 92:102106. [PMID: 39492424 DOI: 10.1016/j.arr.2023.102106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2024]
Abstract
More than a century after the discovery of nicotinamide adenine dinucleotide (NAD+), our understanding of the molecule's role in the biology of ageing continues to evolve. As a coenzyme or substrate for many enzymes, NAD+ governs a wide range of biological processes, including energy metabolism, genomic stability, signal transduction, and cell fate. NAD+ deficiency has been recognised as a bona fide hallmark of tissue degeneration, and restoring NAD+ homeostasis helps to rejuvenate multiple mechanisms associated with tissue ageing. The progressive loss of skeletal muscle homeostasis with age is directly associated with high morbidity, disability and mortality. The aetiology of skeletal muscle ageing is complex, involving mitochondrial dysfunction, senescence and stem cell depletion, autophagy defects, chronic cellular stress, intracellular ion overload, immune cell dysfunction, circadian clock disruption, microcirculation disorders, persistent denervation, and gut microbiota dysbiosis. This review focuses on the therapeutic potential of NAD+ restoration to alleviate the above pathological factors and discusses the effects of in vivo administration of different NAD+ boosting strategies on skeletal muscle homeostasis, aiming to provide a reference for combating skeletal muscle ageing.
Collapse
Affiliation(s)
- Yingying Xu
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Weihua Xiao
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
4
|
Ruskovska T, Bernlohr DA. The Role of NAD + in Metabolic Regulation of Adipose Tissue: Implications for Obesity-Induced Insulin Resistance. Biomedicines 2023; 11:2560. [PMID: 37761000 PMCID: PMC10526756 DOI: 10.3390/biomedicines11092560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Obesity-induced insulin resistance is among the key factors in the development of type 2 diabetes, atherogenic dyslipidemia and cardiovascular disease. Adipose tissue plays a key role in the regulation of whole-body metabolism and insulin sensitivity. In obesity, adipose tissue becomes inflamed and dysfunctional, exhibiting a modified biochemical signature and adipokine secretion pattern that promotes insulin resistance in peripheral tissues. An important hallmark of dysfunctional obese adipose tissue is impaired NAD+/sirtuin signaling. In this chapter, we summarize the evidence for impairment of the NAD+/sirtuin pathway in obesity, not only in white adipose tissue but also in brown adipose tissue and during the process of beiging, together with correlative evidence from human studies. We also describe the role of PARPs and CD38 as important NAD+ consumers and discuss findings from experimental studies that investigated potential NAD+ boosting strategies and their efficacy in restoring impaired NAD+ metabolism in dysfunctional obese adipose tissue. In sum, these studies suggest a critical role of NAD+ metabolism in adipose biology and provide a basis for the potential development of strategies to restore metabolic health in obesity.
Collapse
Affiliation(s)
- Tatjana Ruskovska
- Faculty of Medical Sciences, Goce Delcev University, 2000 Stip, North Macedonia;
| | - David A. Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota—Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
Hepatic Nampt Deficiency Aggravates Dyslipidemia and Fatty Liver in High Fat Diet Fed Mice. Cells 2023; 12:cells12040568. [PMID: 36831235 PMCID: PMC9954480 DOI: 10.3390/cells12040568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/11/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (Nampt) is the rate-limiting enzyme in the salvage pathway of nicotinamide adenine dinucleotide (NAD) biosynthesis. Thus far, hepatic Nampt has not been extensively explored in terms of its effects on serum lipid stability and liver lipids metabolism. In this study, hepatocyte-specific Nampt knockout (HC-Nampt-/-) mice were generated by Cre/loxP system. Nampt mRNA expression was reduced in the liver, but not in other tissues, in HC-Nampt-/- mice compared with wild-type (WT) mice. Hepatic Nampt deficiency had no effect on body weight and fasting blood glucose, and it did not induce atherosclerosis in mice under both normal chow diet (NCD) and high fat diet (HFD). At baseline state under NCD, hepatic Nampt deficiency also did not affect liver weight, liver function index, including alanine aminotransferase, aspartate aminotransferase, albumin and alkaline phosphatase, and serum levels of lipids, including triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and non-esterified fatty acids (NEFA). However, under HFD, deficiency of hepatic Nampt resulted in increased liver weight, liver function index, and serum levels of TG, TC, HDL-C, and NEFA. Meanwhile, histopathological examination showed increased fat accumulation and fibrosis in the liver of HC-Nampt-/- mice compared with WT mice. Taken together, our results show that hepatic Nampt deficiency aggravates dyslipidemia and liver damage in HFD fed mice. Hepatocyte Nampt can be a protective target against dyslipidemia and fatty liver.
Collapse
|
6
|
Extracellular Vesicles in Aging: An Emerging Hallmark? Cells 2023; 12:cells12040527. [PMID: 36831194 PMCID: PMC9954704 DOI: 10.3390/cells12040527] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane-enclosed particles secreted by cells and circulating in body fluids. Initially considered as a tool to dispose of unnecessary material, they are now considered an additional method to transmit cell signals. Aging is characterized by a progressive impairment of the physiological functions of tissues and organs. The causes of aging are complex and interconnected, but there is consensus that genomic instability, telomere erosion, epigenetic alteration, and defective proteostasis are primary hallmarks of the aging process. Recent studies have provided evidence that many of these primary stresses are associated with an increased release of EVs in cell models, able to spread senescence signals in the recipient cell. Additional investigations on the role of EVs during aging also demonstrated the great potential of EVs for the modulation of age-related phenotypes and for pro-rejuvenation therapies, potentially beneficial for many diseases associated with aging. Here we reviewed the current literature on EV secretion in senescent cell models and in old vs. young individual body fluids, as well as recent studies addressing the potential of EVs from different sources as an anti-aging tool. Although this is a recent field, the robust consensus on the altered EV release in aging suggests that altered EV secretion could be considered an emerging hallmark of aging.
Collapse
|
7
|
Ławniczak A, Wrońska A, Wierzbicki P, Kmieć Z. Aging and short-term calorie restriction differently affect the cardiac and skeletal muscle expression of genes regulating energy substrate utilization in male rats. Biogerontology 2022; 23:325-340. [PMID: 35606458 DOI: 10.1007/s10522-022-09965-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/29/2022] [Indexed: 01/11/2023]
Abstract
Aging affects the energy metabolism differently in the cardiac and skeletal muscles. The study aim was to assess the effects of short-term calorie restriction (SCR) and refeeding on the expression of genes involved in the control of cardiac and skeletal muscle energy metabolism in old vs. young male rats. Young (4 mo) and old (24 mo) rats were subjected to 60% SCR for 30 days, and refed ad libitum for 2 or 4 days. In the cardiac (CM) and skeletal muscles (SM) we compared the gene expression (qPCR) of carnitine palmitoyltransferase-I (Cpt-I), peroxisome proliferator-activated receptor beta/delta (Ppar-β/δ), glucose transporter 4 (Glut4), peroxisome proliferator-activated receptor-γ coactivator-1α (Pgc-1α), and sirtuin 3 (Sirt3). In CM, aging increased Cpt-I expression but did not affect the other genes. In SM, Cpt-I, Glut4, Pgc-1α, and Sirt3 mRNA levels were lower in old than young rats. In CM of only young rats SCR increased Cpt-I expression which remained elevated after refeeding. Upon SCR, the expression of Ppar-β/δ, Glut4, Pgc-1α, and Sirt3 in CM increased in young but not old rats, and refeeding re-established control levels. In SM of young rats SCR increased Ppar-β/δ and Pgc-1α, and decreased Sirt3 expression, whereas refeeding generally decreased these mRNA levels. In SM of old rats SCR decreased only Pgc-1α expression. The adaptive response to SCR and subsequent refeeding is muscle tissue-specific and differs in young and old male rats. SCR appears to increase the efficiency of glucose and fatty acid utilization in the cardiac muscle of young, but not old male rats.
Collapse
Affiliation(s)
- Aleksandra Ławniczak
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| | - Agata Wrońska
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland.
| | - Piotr Wierzbicki
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| | - Zbigniew Kmieć
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| |
Collapse
|
8
|
Conlon N, Ford D. A systems-approach to NAD+ restoration. Biochem Pharmacol 2022; 198:114946. [DOI: 10.1016/j.bcp.2022.114946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
9
|
Wang H, Zhu S, Wu X, Liu Y, Ge J, Wang Q, Gu L. NAMPT reduction-induced NAD + insufficiency contributes to the compromised oocyte quality from obese mice. Aging Cell 2021; 20:e13496. [PMID: 34662475 PMCID: PMC8590097 DOI: 10.1111/acel.13496] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/14/2021] [Accepted: 09/12/2021] [Indexed: 12/27/2022] Open
Abstract
Maternal obesity is associated with multiple adverse reproductive outcomes, whereas the underlying molecular mechanisms are still not fully understood. Here, we found the reduced nicotinamide phosphoribosyl transferase (NAMPT) expression and lowered nicotinamide adenine dinucleotide (NAD+) content in oocytes from obese mice. Next, by performing morpholino knockdown assay and pharmacological inhibition, we revealed that NAMPT deficiency not only severely disrupts maturational progression and meiotic apparatus, but also induces the metabolic dysfunction in oocytes. Furthermore, overexpression analysis demonstrated that NAMPT insufficiency induced NAD+ loss contributes to the compromised developmental potential of oocytes and early embryos from obese mice. Importantly, in vitro supplement and in vivo administration of nicotinic acid (NA) was able to ameliorate the obesity‐associated meiotic defects and oxidative stress in oocytes. Our results indicate a role of NAMPT in modulating oocyte meiosis and metabolism, and uncover the beneficial effects of NA treatment on oocyte quality from obese mice.
Collapse
Affiliation(s)
- Hengjie Wang
- College of Animal Science & Technology Nanjing Agricultural University Nanjing China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine Suzhou Municipal Hospital Nanjing Medical University Nanjing China
| | - Xinghan Wu
- Department of Medical Genetics Maternal and Child Health Hospital of Hunan Province Changsha China
| | - Yuan Liu
- College of Animal Science & Technology Nanjing Agricultural University Nanjing China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine Suzhou Municipal Hospital Nanjing Medical University Nanjing China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine Suzhou Municipal Hospital Nanjing Medical University Nanjing China
| | - Ling Gu
- College of Animal Science & Technology Nanjing Agricultural University Nanjing China
| |
Collapse
|
10
|
McReynolds MR, Chellappa K, Chiles E, Jankowski C, Shen Y, Chen L, Descamps HC, Mukherjee S, Bhat YR, Lingala SR, Chu Q, Botolin P, Hayat F, Doke T, Susztak K, Thaiss CA, Lu W, Migaud ME, Su X, Rabinowitz JD, Baur JA. NAD + flux is maintained in aged mice despite lower tissue concentrations. Cell Syst 2021; 12:1160-1172.e4. [PMID: 34559996 DOI: 10.1016/j.cels.2021.09.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 06/08/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022]
Abstract
NAD+ is an essential coenzyme for all living cells. NAD+ concentrations decline with age, but whether this reflects impaired production or accelerated consumption remains unclear. We employed isotope tracing and mass spectrometry to probe age-related changes in NAD+ metabolism across tissues. In aged mice, we observed modest tissue NAD+ depletion (median decrease ∼30%). Circulating NAD+ precursors were not significantly changed, and isotope tracing showed the unimpaired synthesis of nicotinamide from tryptophan. In most tissues of aged mice, turnover of the smaller tissue NAD+ pool was modestly faster such that absolute NAD+ biosynthetic flux was maintained, consistent with more active NAD+-consuming enzymes. Calorie restriction partially mitigated age-associated NAD+ decline by decreasing consumption. Acute inflammatory stress induced by LPS decreased NAD+ by impairing synthesis in both young and aged mice. Thus, the decline in NAD+ with normal aging is relatively subtle and occurs despite maintained NAD+ production, likely due to increased consumption.
Collapse
Affiliation(s)
- Melanie R McReynolds
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Karthikeyani Chellappa
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Chiles
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Connor Jankowski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Yihui Shen
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Li Chen
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Hélène C Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarmistha Mukherjee
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yashaswini R Bhat
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Siddharth R Lingala
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qingwei Chu
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Botolin
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Faisal Hayat
- Department of Pharmacology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Tomohito Doke
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenyun Lu
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Marie E Migaud
- Department of Pharmacology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Xiaoyang Su
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA.
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Saito K, Ito M, Chiba T, Jia H, Kato H. A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions. Nutrients 2021; 13:2277. [PMID: 34209243 PMCID: PMC8308279 DOI: 10.3390/nu13072277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
Many studies have shown the beneficial effects of calorie restriction (CR) on rodents' aging; however, the molecular mechanism explaining these beneficial effects is still not fully understood. Previously, we conducted transcriptomic analysis on rat liver with short-term and mild-to-moderate CR to elucidate its early response to such diet. Here, we expanded transcriptome analysis to muscle, adipose tissue, intestine, and brain and compared the gene expression profiles of these multiple organs and of our previous dataset. Several altered gene expressions were found, some of which known to be related to CR. Notably, the commonly regulated genes by CR include nicotinamide phosphoribosyltransferase and heat shock protein 90, which are involved in declining the aging process and thus potential therapeutic targets for aging-related diseases. The data obtained here provide information on early response markers and key mediators of the CR-induced delay in aging as well as on age-associated pathological changes in mammals.
Collapse
Affiliation(s)
- Kenji Saito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Maiko Ito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Takuya Chiba
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Huijuan Jia
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Hisanori Kato
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| |
Collapse
|
12
|
Pi C, Ma C, Wang H, Sun H, Yu X, Gao X, Yang Y, Sun Y, Zhang H, Shi Y, Li Y, Li Y, He X. MiR-34a suppression targets Nampt to ameliorate bone marrow mesenchymal stem cell senescence by regulating NAD +-Sirt1 pathway. Stem Cell Res Ther 2021; 12:271. [PMID: 33957971 PMCID: PMC8101138 DOI: 10.1186/s13287-021-02339-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Expansion-mediated replicative senescence and age-related natural senescence have adverse effects on mesenchymal stem cell (MSC) regenerative capability and functionality, thus severely impairing the extensive applications of MSC-based therapies. Emerging evidences suggest that microRNA-34a (miR-34a) has been implicated in the process of MSC senescence; however, the molecular mechanisms with regard to how miR-34a influencing MSC senescence remain largely undetermined. METHODS MiR-34a expression in MSCs was evaluated utilizing RT-qPCR. The functional effects of miR-34a exerting on MSC senescence were investigated via gene manipulation. Relevant gene and protein expression levels were analyzed by RT-qPCR and western blot. Luciferase reporter assays were applied to confirm that Nampt is a direct target of miR-34a. The underlying regulatory mechanism of miR-34a targeting Nampt in MSC senescence was further explored by measuring intracellular NAD+ content, NAD+/NADH ratio and Sirt1 activity. RESULTS In contrast to Nampt expression, miR-34a expression incremented in senescent MSCs. MiR-34a overexpression in young MSCs resulted in senescence-associated characteristics as displayed by senescence-like morphology, prolonged cell proliferation, declined osteogenic differentiation potency, heightened senescence-associated-β-galactosidase activity, and upregulated expression levels of the senescence-associated factors. Conversely, miR-34a suppression in replicative senescent and natural senescent MSCs contributed to diminished senescence-related phenotypic features. We identified Nampt as a direct target gene of miR-34a. In addition, miR-34a repletion resulted in prominent reductions in Nampt expression levels, NAD+ content, NAD+/NADH ratio, and Sirt1 activity, whereas anti-miR-34a treatment exerted the opposite effects. Furthermore, miR-34a-mediated MSC senescence was evidently rescued following the co-treatment with Nampt overexpression. CONCLUSION This study identifies a significant role of miR-34a playing in MSC replicative senescence and natural senescence via targeting Nampt and further mediating by NAD+-Sirt1 pathway, carrying great implications for optimal strategies for MSC therapeutic applications.
Collapse
Affiliation(s)
- Chenchen Pi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China.,The First Hospital, and Institute of Immunology, Jilin University, Changchun, 130021, China
| | - Cao Ma
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Huan Wang
- Department of Pathology, The First Affiliated Hospital, Henan University of Chinese Medicine, Henan, 450000, China
| | - Hui Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Xiao Yu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Xingyu Gao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Yue Yang
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Yanan Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Haiying Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Yingai Shi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Yan Li
- Division of Orthopedics and Biotechnology, Department for Clinical Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, Jilin Province, People's Republic of China.
| |
Collapse
|
13
|
Okabe K, Nawaz A, Nishida Y, Yaku K, Usui I, Tobe K, Nakagawa T. NAD+ Metabolism Regulates Preadipocyte Differentiation by Enhancing α-Ketoglutarate-Mediated Histone H3K9 Demethylation at the PPARγ Promoter. Front Cell Dev Biol 2020; 8:586179. [PMID: 33330464 PMCID: PMC7732485 DOI: 10.3389/fcell.2020.586179] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/03/2020] [Indexed: 01/07/2023] Open
Abstract
Obesity has become a serious problem in public health worldwide, causing numerous metabolic diseases. Once the differentiation to mature adipocytes is disrupted, adipocyte hypertrophy and ectopic lipid accumulation leads to the inflammation in adipose tissue and systemic metabolic disorders. Intracellular metabolic state is known to change during cell differentiation and it affects the cell fate or the differentiation through epigenetic mechanism. Although the mechanism of preadipocyte differentiation has been well established, it is unknown how metabolic state changes and how it affects the differentiation in predipocyte differentiation. Nicotinamide adenine dinucleotide (NAD+) plays crucial roles in energy metabolism as a coenzyme in multiple redox reactions in major catabolic pathways and as a substrate of sirtuins or poly(ADP-ribose)polymerases. NAD+ is mainly synthesized from salvage pathway mediated by two enzymes, Nampt and Nmnat. The manipulation to NAD+ metabolism causes metabolic change in each tissue and changes in systemic metabolism. However, the role of NAD+ and Nampt in adipocyte differentiation remains unknown. In this study, we employed liquid chromatography-mass spectrometry (LC-MS)- and gas chromatography-mass spectrometry (GC-MS)-based targeted metabolomics to elucidate the metabolic reprogramming events that occur during 3T3-L1 preadipocyte differentiation. We found that the tricarboxylic acid (TCA) cycle was enhanced, which correlated with upregulated NAD+ synthesis. Additionally, increased alpha-ketoglutarate (αKG) contributed to histone H3K9 demethylation in the promoter region of PPARγ, leading to its transcriptional activation. Thus, we concluded that NAD+-centered metabolic reprogramming is necessary for the differentiation of 3T3-L1 preadipocytes.
Collapse
Affiliation(s)
- Keisuke Okabe
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan.,First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Allah Nawaz
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan.,First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Yasuhiro Nishida
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Isao Usui
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Tochigi, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan.,Research Center for Pre-Disease Science, University of Toyama, Toyama, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan.,Research Center for Pre-Disease Science, University of Toyama, Toyama, Japan
| |
Collapse
|
14
|
Polyakova YV, Zavodovsky BV, Sivordova LE, Akhverdyan YR, Zborovskaya IA. Visfatin and Rheumatoid Arthritis: Pathogenetic Implications and Clinical Utility. Curr Rheumatol Rev 2020; 16:224-239. [DOI: 10.2174/1573397115666190409112621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Objective:
Analysis and generalization of data related to visfatin involvement in the
pathogenesis of inflammation at various stages of rheumatoid arthritis.
Data Synthesis:
Visfatin is an adipocytokine which has also been identified in non-adipose tissues.
It influences directly on the maturation of B cells, which are involved in autoantibody production
and T cell activation. Visfatin can promote inflammation via regulation of pro-inflammatory cytokines
including TNF, IL-1β and IL-6. The concentration of circulating visfatin in rheumatoid arthritis
patients is higher compared to healthy individuals. Several studies suggest that visfatin level is
associated with rheumatoid arthritis activity, and its elevation may precede clinical signs of the relapse.
In murine collagen-induced arthritis, visfatin levels were also found to be elevated both in
inflamed synovial cells and in joint vasculature. Visfatin blockers have been shown to confer fast
and long-term attenuation of pathological processes; however, most of their effects are transient.
Other factors responsible for hyperactivation of the immune system can participate in this process
at a later stage. Treatment of rheumatoid arthritis with a combination of these blockers and inhibitors
of other mediators of inflammation can potentially improve treatment outcomes compared to
current therapeutic strategies. Recent advances in the treatment of experimental arthritis in mice as
well as the application of emerging treatment strategies obtained from oncology for rheumatoid arthritis
management could be a source of novel adipokine-mediated anti-rheumatic drugs.
Conclusion:
The ongoing surge of interest in anticytokine therapy makes further study of visfatin
highly relevant as it may serve as a base for innovational RA treatment.
Collapse
Affiliation(s)
- Yulia V. Polyakova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Boris V. Zavodovsky
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Larisa E. Sivordova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Yuri R. Akhverdyan
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Irina A. Zborovskaya
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| |
Collapse
|
15
|
Zhang Z, Xu HN, Li S, Jr AD, Chellappa K, Davis JG, Guan Y, Frederick DW, Chu W, Zhao H, Li LZ, Baur JA. Rapamycin maintains NAD +/NADH redox homeostasis in muscle cells. Aging (Albany NY) 2020; 12:17786-17799. [PMID: 32960787 PMCID: PMC7585102 DOI: 10.18632/aging.103954] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/03/2020] [Indexed: 01/24/2023]
Abstract
Rapamycin delays multiple age-related conditions and extends lifespan in organisms ranging from yeast to mice. However, the mechanisms by which rapamycin influences longevity are incompletely understood. The objective of this study was to investigate the effect of rapamycin on NAD+/NADH redox balance. We report that the NAD+/NADH ratio of C2C12 myoblasts or differentiated myotubes significantly decreases over time in culture, and that rapamycin prevents this effect. Despite lowering the NADH available to support ATP generation, rapamycin increases ATP availability, consistent with lowering energetic demand. Although rapamycin did not change the NAD+/NADH ratio or steady-state ATP concentration in the livers, kidneys, or muscles of young mice, optical redox imaging revealed that rapamycin caused a substantial decline in the NADH content and an increase in the optical redox ratio (a surrogate of NAD+/NADH redox ratio) in muscles from aged mice. Collectively, these data suggest that rapamycin favors a more oxidized NAD+/NADH ratio in aged muscle, which may influence metabolism and the activity of NAD+-dependent enzymes. This study provides new insight into the mechanisms by which rapamycin might influence the aging process to improve health and longevity among the aging population.
Collapse
Affiliation(s)
- Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China,Institute for Diabetes, Obesity, and Metabolism, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - He N. Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Antonio Davila Jr
- Institute for Diabetes, Obesity, and Metabolism, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karthikeyani Chellappa
- Institute for Diabetes, Obesity, and Metabolism, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James G. Davis
- Institute for Diabetes, Obesity, and Metabolism, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuxia Guan
- Division of Trauma, Critical Care, and Emergency Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David W. Frederick
- Institute for Diabetes, Obesity, and Metabolism, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Weiqing Chu
- Institute for Diabetes, Obesity, and Metabolism, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Huaqing Zhao
- Department of Clinical Sciences, Temple University School of Medicine, Philadelphia, PA 19104, USA
| | - Lin Z. Li
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph A. Baur
- Institute for Diabetes, Obesity, and Metabolism, Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Valayer S, Kim D, Fogtman A, Straube U, Winnard A, Caplan N, Green DA, van Leeuwen FHP, Weber T. The Potential of Fasting and Caloric Restriction to Mitigate Radiation Damage-A Systematic Review. Front Nutr 2020; 7:584543. [PMID: 33072801 PMCID: PMC7530334 DOI: 10.3389/fnut.2020.584543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
Detrimental health effects from ionizing radiation to living organisms is one of the key concerns identified and addressed by Radiation Protection institutions, nationally and internationally on Earth and for human spaceflight. Thus, new methods for mitigating the adverse effects of ionizing radiation are urgently needed for terrestrial health and deep space exploration. Caloric restriction and (intermittent-) fasting have been reported to elicit a variety of immediate and long-term physiological effects. The rapidly growing body of evidence of research studies investigating the effects of caloric restriction and dietary fasting points toward a multitude of benefits affecting numerous physiological systems. Therefore, a systematic review was performed to evaluate the evidence of caloric restriction and dietary fasting on the physiological response to ionizing radiation in humans and animals. All experimental studies of humans, animals, and eukaryotic cell lines available in PubMed, Cochrane library, and specialized databases were searched comparing irradiation post-caloric restriction or fasting to a non-nutritionally restricted control group on a broad range of outcomes from molecular to clinical responses. The initial search yielded 2,653 records. The final analysis included 11 studies. Most studies investigated survival rate or cancer occurrence in animals. Included studies did not reveal any benefit from pre exposure caloric restriction, except when performed with post radiation caloric restriction. However, the effects of pre-exposure fasting suggest increased resilience to ionizing radiation.
Collapse
Affiliation(s)
- Simon Valayer
- European Space Agency (ESA), European Astronaut Center (EAC), Space Medicine Team (HRE-OM), Cologne, Germany.,Faculty of Medicine Paris VI, Sorbonne University, Paris, France
| | - David Kim
- European Space Agency (ESA), European Astronaut Center (EAC), Space Medicine Team (HRE-OM), Cologne, Germany.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Anna Fogtman
- European Space Agency (ESA), European Astronaut Center (EAC), Space Medicine Team (HRE-OM), Cologne, Germany
| | - Ulrich Straube
- European Space Agency (ESA), European Astronaut Center (EAC), Space Medicine Team (HRE-OM), Cologne, Germany
| | - Andrew Winnard
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Nick Caplan
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - David A Green
- European Space Agency (ESA), European Astronaut Center (EAC), Space Medicine Team (HRE-OM), Cologne, Germany.,KBR GmbH, Cologne, Germany.,Center of Human & Applied Physiological Sciences (CHAPS), King's College London, London, United Kingdom
| | - Flora H P van Leeuwen
- European Space Agency (ESA), European Astronaut Center (EAC), Space Medicine Team (HRE-OM), Cologne, Germany.,Faculty of Medicine, Utrecht University, Utrecht, Netherlands
| | - Tobias Weber
- European Space Agency (ESA), European Astronaut Center (EAC), Space Medicine Team (HRE-OM), Cologne, Germany.,KBR GmbH, Cologne, Germany
| |
Collapse
|
17
|
Pi C, Yang Y, Sun Y, Wang H, Sun H, Ma M, Lin L, Shi Y, Li Y, Li Y, He X. Nicotinamide phosphoribosyltransferase postpones rat bone marrow mesenchymal stem cell senescence by mediating NAD +-Sirt1 signaling. Aging (Albany NY) 2020; 11:3505-3522. [PMID: 31175267 PMCID: PMC6594813 DOI: 10.18632/aging.101993] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 05/22/2019] [Indexed: 12/25/2022]
Abstract
In vitro replicative senescence affects MSC characteristics and functionality, thus severely restricting their application in regenerative medicine and MSC-based therapies. Previously, we found that MSC natural senescence is accompanied by altered intracellular nicotinamide adenine dinucleotide (NAD+) metabolism, in which Nampt plays a key role. However, whether Nampt influences MSC replicative senescence is still unclear. Our study showed that Nampt expression is down-regulated during MSC replicative senescence. Nampt depletion via a specific Nampt inhibitor FK866 or Nampt knockdown in early passage MSCs led to enhanced senescence as indicated by senescence-like morphology, reduced proliferation, and adipogenic and osteogenic differentiation, and increased senescence-associated-β-galactosidase activity and the expression of the senescence-associated factor p16INK4a. Conversely, Nampt overexpression ameliorated senescence-associated phenotypic features in late passage MSCs. Further, Nampt inhibition resulted in reduced intracellular NAD+ content, NAD+/NADH ratio, and Sirt1 activity, whereas overexpression had the opposite effects. Exogenous intermediates involved in NAD+ biosynthesis not only rescued replicative senescent MSCs but also alleviated FK866-induced MSC senescence. Thus, Nampt suppresses MSC senescence via mediating NAD+-Sirt1 signaling. This study provides novel mechanistic insights into MSC replicative senescence and a promising strategy for the severe shortage of cells for MSC-based therapies.
Collapse
Affiliation(s)
- Chenchen Pi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.,The First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Yue Yang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.,Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Yanan Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Huan Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Hui Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Mao Ma
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.,Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lin Lin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yingai Shi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yan Li
- Division of Orthopedics and Biotechnology, Department for Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
18
|
Pardo PS, Boriek AM. SIRT1 Regulation in Ageing and Obesity. Mech Ageing Dev 2020; 188:111249. [PMID: 32320732 DOI: 10.1016/j.mad.2020.111249] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 03/12/2020] [Accepted: 04/05/2020] [Indexed: 12/29/2022]
Abstract
Ageing and obesity have common hallmarks: altered glucose and lipid metabolism, chronic inflammation and oxidative stress are some examples. The downstream effects of SIRT1 activity have been thoroughly explored, and their research is still in expanse. SIRT1 activation has been shown to regulate pathways with beneficiary effects on 1) ageing and obesity-associated metabolic disorders such as metabolic syndrome, insulin resistance and type-II diabetes with, 2) chronic inflammatory processes such as arthritis, atherosclerosis and emphysema, 3) DNA damage and oxidative stress with impact on neurodegenerative diseases, cardiovascular health and some cancers. This knowledge intensified the interest in uncovering the mechanisms regulating the expression and activity of SIRT1. This review focuses on the upstream regulatory mechanisms controlling SIRT1, and how this knowledge could potentially contribute to the development of therapeutic interventions.
Collapse
Affiliation(s)
- Patricia S Pardo
- Pulmonary and Critical Care medicine, Department of Medicine, Baylor College of Medicine, Houston TX 77030, USA.
| | - Aladin M Boriek
- Pulmonary and Critical Care medicine, Department of Medicine, Baylor College of Medicine, Houston TX 77030, USA.
| |
Collapse
|
19
|
Wei X, Jia R, Wang G, Hong S, Song L, Sun B, Chen K, Wang N, Wang Q, Luo X, Yan J. Depot-specific regulation of NAD +/SIRTs metabolism identified in adipose tissue of mice in response to high-fat diet feeding or calorie restriction. J Nutr Biochem 2020; 80:108377. [PMID: 32278117 DOI: 10.1016/j.jnutbio.2020.108377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 12/31/2022]
Abstract
Deteriorated nicotinamide adenine dinucleotide (NAD+)/sirtuins (SIRTs) metabolism in adipose tissue is implicated in diet-induced obesity, while calorie restriction (CR)-induced beneficial effects require sufficient NAD+ biosynthesis. Mechanistic links have not been defined. This study aims to identify changes of specific components of NAD+/SIRTs system in white adipose tissue (WAT) and brown adipose tissue (BAT) of mice upon energy imbalance, focusing on key enzymes in NAD+ salvage (Nampt, Nmnat1, Nrk1), clearance (Nnmt, Aox1, Cyp2e1) and consumption pathways (Sirt1, Sirt2, Sirt3, Sirt6, Parp1). Male C57BL/6J mice were fed ad libitum with the standard laboratory chow diet, high-fat diet (HFD) or 40% CR diet, respectively. The epididymal and inguinal WAT (eWAT and iWAT) and interscapular BAT (iBAT) were harvested for histological, NAD+ assay, gene and protein expression analysis after 16 weeks of dietary regimen. HFD decreased, while CR increased, the NAD+ and NADH levels in eWAT, iWAT and iBAT. NAD+ content negatively correlated with plasma cholesterol, TNF-α levels and calorie intake, while it positively correlated with plasma adiponectin level. The change trend of SIRT1 is quite the same as that of NAD+/NADH ratio. Nmnat1 gene is sensitive to energy imbalance in WAT but not in BAT. Nrk1 gene expression was decreased in eWAT and iWAT but increased in iBAT of HFD mice. Nnmt mRNA and protein abundance was increased in iWAT of HFD mice. Nampt, Cyp2e1 and Sirt3 were the most robust genes responding to energy imbalance. In summary, adipose tissue responds to long-term energy excess or shortage with depot-specific transcriptional activation or repression of NAD+/SIRTs metabolic components.
Collapse
Affiliation(s)
- Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Ru Jia
- Department of Prosthodontics, College of Stomatology, Stomatological Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Guan Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering and School of Life Sciences, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China
| | - Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Ke Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Nan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Qiqi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
| |
Collapse
|
20
|
Dollerup OL, Chubanava S, Agerholm M, Søndergård SD, Altıntaş A, Møller AB, Høyer KF, Ringgaard S, Stødkilde-Jørgensen H, Lavery GG, Barrès R, Larsen S, Prats C, Jessen N, Treebak JT. Nicotinamide riboside does not alter mitochondrial respiration, content or morphology in skeletal muscle from obese and insulin-resistant men. J Physiol 2019; 598:731-754. [PMID: 31710095 DOI: 10.1113/jp278752] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/01/2019] [Indexed: 01/02/2023] Open
Abstract
KEY POINTS This is the first long-term human clinical trial to report on effects of nicotinamide riboside (NR) on skeletal muscle mitochondrial function, content and morphology. NR supplementation decreases nicotinamide phosphoribosyltransferase (NAMPT) protein abundance in skeletal muscle. NR supplementation does not affect NAD metabolite concentrations in skeletal muscle. Respiration, distribution and quantity of muscle mitochondria are unaffected by NR. NAMPT in skeletal muscle correlates positively with oxidative phosphorylation Complex I, sirtuin 3 and succinate dehydrogenase. ABSTRACT Preclinical evidence suggests that the nicotinamide adenine dinucleotide (NAD+ ) precursor nicotinamide riboside (NR) boosts NAD+ levels and improves diseases associated with mitochondrial dysfunction. We aimed to determine if dietary NR supplementation in middle-aged, obese, insulin-resistant men affects mitochondrial respiration, content and morphology in skeletal muscle. In a randomized, placebo-controlled clinical trial, 40 participants received 1000 mg NR or placebo twice daily for 12 weeks. Skeletal muscle biopsies were collected before and after the intervention. Mitochondrial respiratory capacity was determined by high-resolution respirometry on single muscle fibres. Protein abundance and mRNA expression were measured by Western blot and quantitative PCR analyses, respectively, and in a subset of the participants (placebo n = 8; NR n = 8) we quantified mitochondrial fractional area and mitochondrial morphology by laser scanning confocal microscopy. Protein levels of nicotinamide phosphoribosyltransferase (NAMPT), an essential NAD+ biosynthetic enzyme in skeletal muscle, decreased by 14% with NR. However, steady-state NAD+ levels as well as gene expression and protein abundance of other NAD+ biosynthetic enzymes remained unchanged. Neither respiratory capacity of skeletal muscle mitochondria nor abundance of mitochondrial associated proteins were affected by NR. Moreover, no changes in mitochondrial fractional area or network morphology were observed. Our data do not support the hypothesis that dietary NR supplementation has significant impact on skeletal muscle mitochondria in obese and insulin-resistant men. Future studies on the effects of NR on human skeletal muscle may include both sexes and potentially provide comparisons between young and older people.
Collapse
Affiliation(s)
- Ole L Dollerup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Medical Research Laboratory, Department of Clinical Medicine, Aarhus University Hospital, Denmark.,Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Denmark.,Department of Clinical Pharmacology, Aarhus University Hospital, Denmark
| | - Sabina Chubanava
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Marianne Agerholm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Stine D Søndergård
- Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Ali Altıntaş
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Andreas B Møller
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark
| | - Kasper F Høyer
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Denmark.,The MR Research Centre, Aarhus University Hospital, Denmark
| | | | | | - Gareth G Lavery
- Clinical and Experimental Medicine, University of Birmingham, UK
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Steen Larsen
- Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Denmark.,Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Clara Prats
- Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Niels Jessen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Denmark.,Department of Clinical Pharmacology, Aarhus University Hospital, Denmark.,Department of Biomedicine, Aarhus University, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
21
|
Mitani T, Watanabe S, Wada K, Fujii H, Nakamura S, Katayama S. Intracellular cAMP contents regulate NAMPT expression via induction of C/EBPβ in adipocytes. Biochem Biophys Res Commun 2019; 522:770-775. [PMID: 31791580 DOI: 10.1016/j.bbrc.2019.11.165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/25/2019] [Indexed: 11/28/2022]
Abstract
A decline in intracellular nicotinamide adenine mononucleotide (NAD+) causes adipose tissue dysfunction. Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step in the NAD+ biosynthesis pathway. However, the molecular mechanism that mediates regulation of NAMPT expression in adipocytes is yet to be elucidated. This study found that intracellular cAMP regulates NAMPT expression and promoter activity in 3T3-L1 adipocytes. cAMP-mediated Nampt promoter activity was suppressed by protein kinase A inhibitor H89, whereas AMP-activated protein kinase inhibitor compound C did not affect cAMP-mediated Nampt promoter activity. Intracellular cAMP induced CCAAT/enhancer-binding protein β (C/EBPβ) expression. Knockdown of C/EBPβ suppressed NAMPT expression and promoter activity. Furthermore, the Nampt promoter was activated by C/EBPβ, while LIP activated the dominant-negative form of C/EBPβ. Promoter sequence analysis revealed that the region from -96 to -76 on Nampt was required for C/EBPβ-mediated promoter activity. Additionally, chromatin immunoprecipitation assay demonstrated that C/EBPβ was bound to the promoter sequences of Nampt. Finally, NAMPT inhibitor FK866 suppressed adipogenesis in 3T3-L1 cells, and this suppressive effect was restored by nicotinamide mononucleotide treatment. These findings showed that intracellular cAMP increased NAMPT levels by induction of C/EBPβ expression and indicated that the induction of NAMPT expression was important for adipogenesis.
Collapse
Affiliation(s)
- Takakazu Mitani
- Graduate School of Science and Technology, Department of Agriculture, Division of Food Science and Biotechnology, Shinshu University, Kami-ina, Nagano, Japan.
| | - Shun Watanabe
- Graduate School of Science and Technology, Department of Agriculture, Division of Food Science and Biotechnology, Shinshu University, Kami-ina, Nagano, Japan
| | - Kenjiro Wada
- Department of Bioscience and Biotechnology, Shinshu University, Kami-ina, Nagano, Japan
| | - Hiroshi Fujii
- Graduate School of Science and Technology, Department of Biomedical Engineering, Shinshu University, Kami-ina, Nagano, Japan; Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Kami-ina, Nagano, Japan
| | - Soichiro Nakamura
- Graduate School of Science and Technology, Department of Agriculture, Division of Food Science and Biotechnology, Shinshu University, Kami-ina, Nagano, Japan
| | - Shigeru Katayama
- Graduate School of Science and Technology, Department of Agriculture, Division of Food Science and Biotechnology, Shinshu University, Kami-ina, Nagano, Japan; Department of Biomolecular Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Kami-ina, Nagano, Japan
| |
Collapse
|
22
|
The Novel Perspectives of Adipokines on Brain Health. Int J Mol Sci 2019; 20:ijms20225638. [PMID: 31718027 PMCID: PMC6887733 DOI: 10.3390/ijms20225638] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
First seen as a fat-storage tissue, the adipose tissue is considered as a critical player in the endocrine system. Precisely, adipose tissue can produce an array of bioactive factors, including cytokines, lipids, and extracellular vesicles, which target various systemic organ systems to regulate metabolism, homeostasis, and immune response. The global effects of adipokines on metabolic events are well defined, but their impacts on brain function and pathology remain poorly defined. Receptors of adipokines are widely expressed in the brain. Mounting evidence has shown that leptin and adiponectin can cross the blood–brain barrier, while evidence for newly identified adipokines is limited. Significantly, adipocyte secretion is liable to nutritional and metabolic states, where defective circuitry, impaired neuroplasticity, and elevated neuroinflammation are symptomatic. Essentially, neurotrophic and anti-inflammatory properties of adipokines underlie their neuroprotective roles in neurodegenerative diseases. Besides, adipocyte-secreted lipids in the bloodstream can act endocrine on the distant organs. In this article, we have reviewed five adipokines (leptin, adiponectin, chemerin, apelin, visfatin) and two lipokines (palmitoleic acid and lysophosphatidic acid) on their roles involving in eating behavior, neurotrophic and neuroprotective factors in the brain. Understanding and regulating these adipokines can lead to novel therapeutic strategies to counteract metabolic associated eating disorders and neurodegenerative diseases, thus promote brain health.
Collapse
|
23
|
Csiszar A, Tarantini S, Yabluchanskiy A, Balasubramanian P, Kiss T, Farkas E, Baur JA, Ungvari Z. Role of endothelial NAD + deficiency in age-related vascular dysfunction. Am J Physiol Heart Circ Physiol 2019; 316:H1253-H1266. [PMID: 30875255 PMCID: PMC6620681 DOI: 10.1152/ajpheart.00039.2019] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/26/2019] [Accepted: 03/12/2019] [Indexed: 12/23/2022]
Abstract
Age-related alterations in endothelium and the resulting vascular dysfunction critically contribute to a range of pathological conditions associated with old age. To develop therapies rationally that improve vascular health and thereby increase health span and life span in older adults, it will be essential to understand the cellular and molecular mechanisms contributing to vascular aging. Preclinical studies in model organisms demonstrate that NAD+ availability decreases with age in multiple tissues and that supplemental NAD+ precursors can ameliorate many age-related cellular impairments. Here, we provide a comprehensive overview of NAD+-dependent pathways [including the NAD+-using silent information regulator-2-like enzymes and poly(ADP-ribose) polymerase enzymes] and the potential consequences of endothelial NAD+ deficiency in vascular aging. The multifaceted vasoprotective effects of treatments that reverse the age-related decline in cellular NAD+ levels, as well as their potential limitations, are discussed. The preventive and therapeutic potential of NAD+ intermediates as effective, clinically relevant interventions in older adults at risk for ischemic heart disease, vascular cognitive impairment, and other common geriatric conditions and diseases that involve vascular pathologies (e.g., sarcopenia, frailty) are critically discussed. We propose that NAD+ precursors [e.g., nicotinamide (Nam) riboside, Nam mononucleotide, niacin] should be considered as critical components of combination therapies to slow the vascular aging process and increase cardiovascular health span.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
- Theoretical Medicine Doctoral School, University of Szeged , Szeged , Hungary
| | - Eszter Farkas
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
- Theoretical Medicine Doctoral School, University of Szeged , Szeged , Hungary
- Department of Pulmonology, Semmelweis University , Budapest , Hungary
- Department of Health Promotion Sciences, Hudson College of Public Health, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| |
Collapse
|
24
|
Calderón-Torres CM, Sarabia-Curz L, Ledesma-Soto Y, Murguía-Romero M, Terrazas LI. Denitrase activity of Debaryomyces hansenii reduces the oxidized compound 3-nitrotyrosine in mice liver with colitis. Exp Ther Med 2019; 17:3748-3754. [PMID: 31007730 DOI: 10.3892/etm.2019.7395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 02/20/2019] [Indexed: 12/15/2022] Open
Abstract
The oxidation of tyrosine to 3-nitrotyrosine is irreversible, and due to this characteristic, 3-nitrotyrosine is used as a marker for oxidative stress in a range of diverse chronic and degenerative diseases. It has been established that the yeast Debaryomyces hansenii (D. hansenii) can assimilate free 3-nitrotyrosine as unique source of nitrogen, and during saline stress, has a high denitrase activity to detoxify this compound in a reaction that involves the liberation of nitrogen dioxide from 3-nitrotyrosine. However, until now it has not been determined whether D. hansenii can detoxify protein-bound 3-nitrotyrosine such as nitrated proteins present in different chronic illnesses. TThe aim of the present study was to evaluate the denitrase activity of D. hansenii to reduce 3-nitrotyrosine from liver proteins of mice with colitis. Firstly, the levels of reactive oxygen species of liver tissue of colitic and control mice were measured by the reaction with the 2'7'-dichlorofluorescein diacetate. Denitrase activity of D. hansenii was evaluated by incubating cell extracts of the yeast with protein extracts from livers of mice with colitis. Following incubation, 3-nitrotyrosine was measured, and to corroborate that denitrase reaction had occurred, the production of nitrites was measured. In samples of liver tissue from mice with colitis, the maximum levels of reactive oxygen species were up to two times higher compared with the control livers. Following the incubation of colitic liver samples with cell extracts of D. hansenii, it was observed that 3-nitrotyrosine decreased to the basal concentration of control liver samples, and that the concentration of nitrites was increased. These results indicate that denitrase of D. hansenii extracts can effectively detoxify 3-nitrotyrosine bound to proteins and that the extracts could be used to decrease protein oxidation damage in chronic degenerative diseases.
Collapse
Affiliation(s)
| | - Lirio Sarabia-Curz
- Biomedicine Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Tlalnepantla 54090, Mexico
| | - Yadira Ledesma-Soto
- Biomedicine Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Tlalnepantla 54090, Mexico
| | - Miguel Murguía-Romero
- Morphology and Function Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Tlalnepantla 54090, Mexico
| | - Luis I Terrazas
- Biomedicine Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Tlalnepantla 54090, Mexico
| |
Collapse
|
25
|
Hoizumi M, Sato T, Shimizu T, Kato S, Tsukiyama K, Narita T, Fujita H, Morii T, Sassa MH, Seino Y, Yamada Y. Inhibition of GIP signaling extends lifespan without caloric restriction. Biochem Biophys Res Commun 2019; 513:974-982. [PMID: 31003779 DOI: 10.1016/j.bbrc.2019.04.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/04/2019] [Indexed: 01/02/2023]
Abstract
AIMS/INTRODUCTION Caloric restriction (CR) promotes longevity and exerts anti-aging effects by increasing Sirtuin production and activation. Gastric inhibitory polypeptide (GIP), a gastrointestinal peptide hormone, exerts various effects on pancreatic β-cells and extra-pancreatic tissues. GIP promotes glucose-dependent augmentation of insulin secretion and uptake of nutrients into the adipose tissue. MATERIALS AND METHODS Gipr-/- and Gipr+/+ mice were used for lifespan analysis, behavior experiments and gene expression of adipose tissue and muscles. 3T3-L1 differentiated adipocytes were used for Sirt1 and Nampt expression followed by treatment with GIP and α-lipoic acid. RESULTS We observed that GIP receptor-knockout (Gipr-/-) mice fed normal diet showed an extended lifespan, increased exploratory and decreased anxiety-based behaviors, which are characteristic behavioral changes under CR. Moreover, Gipr-/- mice showed increased Sirt1 and Nampt expression in the adipose tissue. GIP suppressed α-lipoic acid-induced Sirt1 expression and activity in differentiated adipocytes. CONCLUSIONS Although maintenance of CR is difficult, food intake and muscle endurance of Gipr-/- mice were similar to those of wild-type mice. Inhibition of GIP signaling may be a novel strategy to extend the lifespan of diabetic patients.
Collapse
Affiliation(s)
- Manabu Hoizumi
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Takehiro Sato
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Tatsunori Shimizu
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Shunsuke Kato
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Katsushi Tsukiyama
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Takuma Narita
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Hiroki Fujita
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Tsukasa Morii
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan
| | - Mariko Harada Sassa
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Japan
| | - Yutaka Seino
- Kansai Electric Power Medical Research Institute, Osaka, Japan
| | - Yuichiro Yamada
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Japan.
| |
Collapse
|
26
|
Kobayashi M, Fujii N, Narita T, Higami Y. SREBP-1c-Dependent Metabolic Remodeling of White Adipose Tissue by Caloric Restriction. Int J Mol Sci 2018; 19:ijms19113335. [PMID: 30373107 PMCID: PMC6275055 DOI: 10.3390/ijms19113335] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/21/2018] [Accepted: 10/21/2018] [Indexed: 12/21/2022] Open
Abstract
Caloric restriction (CR) delays the onset of many age-related pathophysiological changes and extends lifespan. White adipose tissue (WAT) is not only a major tissue for energy storage, but also an endocrine tissue that secretes various adipokines. Recent reports have demonstrated that alterations in the characteristics of WAT can impact whole-body metabolism and lifespan. Hence, we hypothesized that functional alterations in WAT may play important roles in the beneficial effects of CR. Previously, using microarray analysis of WAT from CR rats, we found that CR enhances fatty acid (FA) biosynthesis, and identified sterol regulatory element-binding protein 1c (SREBP-1c), a master regulator of FA synthesis, as a mediator of CR. These findings were validated by showing that CR failed to upregulate factors involved in FA biosynthesis and to extend longevity in SREBP-1c knockout mice. Furthermore, we revealed that SREBP-1c is implicated in CR-associated mitochondrial activation through the upregulation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis. Notably, these CR-associated phenotypes were observed only in WAT. We conclude that CR induces SREBP-1c-dependent metabolic remodeling, including the enhancement of FA biosynthesis and mitochondrial activation, via PGC-1α in WAT, resulting in beneficial effects.
Collapse
Affiliation(s)
- Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
- Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| | - Namiki Fujii
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| | - Takumi Narita
- Epidemiology and Prevention Division, Research Center for Cancer Prevention and Screening, National Cancer Center, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
- Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan.
| |
Collapse
|
27
|
Li D, Ke Y, Zhan R, Liu C, Zhao M, Zeng A, Shi X, Ji L, Cheng S, Pan B, Zheng L, Hong H. Trimethylamine-N-oxide promotes brain aging and cognitive impairment in mice. Aging Cell 2018; 17:e12768. [PMID: 29749694 PMCID: PMC6052480 DOI: 10.1111/acel.12768] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2018] [Indexed: 12/21/2022] Open
Abstract
Gut microbiota can influence the aging process and may modulate aging‐related changes in cognitive function. Trimethylamine‐N‐oxide (TMAO), a metabolite of intestinal flora, has been shown to be closely associated with cardiovascular disease and other diseases. However, the relationship between TMAO and aging, especially brain aging, has not been fully elucidated. To explore the relationship between TMAO and brain aging, we analysed the plasma levels of TMAO in both humans and mice and administered exogenous TMAO to 24‐week‐old senescence‐accelerated prone mouse strain 8 (SAMP8) and age‐matched senescence‐accelerated mouse resistant 1 (SAMR1) mice for 16 weeks. We found that the plasma levels of TMAO increased in both the elderly and the aged mice. Compared with SAMR1‐control mice, SAMP8‐control mice exhibited a brain aging phenotype characterized by more senescent cells in the hippocampal CA3 region and cognitive dysfunction. Surprisingly, TMAO treatment increased the number of senescent cells, which were primarily neurons, and enhanced the mitochondrial impairments and superoxide production. Moreover, we observed that TMAO treatment increased synaptic damage and reduced the expression levels of synaptic plasticity‐related proteins by inhibiting the mTOR signalling pathway, which induces and aggravates aging‐related cognitive dysfunction in SAMR1 and SAMP8 mice, respectively. Our findings suggested that TMAO could induce brain aging and age‐related cognitive dysfunction in SAMR1 mice and aggravate the cerebral aging process of SAMP8 mice, which might provide new insight into the effects of intestinal microbiota on the brain aging process and help to delay senescence by regulating intestinal flora metabolites.
Collapse
Affiliation(s)
- Dang Li
- Department of Geriatrics; Fujian Medical University Union Hospital; Fuzhou China
| | - Yilang Ke
- Department of Geriatrics; Fujian Medical University Union Hospital; Fuzhou China
| | - Rui Zhan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine; School of Basic Medical Sciences; Peking University Health Science Center; Key Laboratory of Molecular Cardiovascular Science; Ministry of Education; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Ministry of Health; Beijing Key Laboratory of Cardiovascular Receptors Research; Beijing China
| | - Changjie Liu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine; School of Basic Medical Sciences; Peking University Health Science Center; Key Laboratory of Molecular Cardiovascular Science; Ministry of Education; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Ministry of Health; Beijing Key Laboratory of Cardiovascular Receptors Research; Beijing China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine; School of Basic Medical Sciences; Peking University Health Science Center; Key Laboratory of Molecular Cardiovascular Science; Ministry of Education; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Ministry of Health; Beijing Key Laboratory of Cardiovascular Receptors Research; Beijing China
| | - Aiping Zeng
- Department of Cardiology; Fujian Medical University Union Hospital; Fuzhou China
| | - Xiaoyun Shi
- Department of Geriatrics; Fujian Medical University Union Hospital; Fuzhou China
| | - Liang Ji
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine; School of Basic Medical Sciences; Peking University Health Science Center; Key Laboratory of Molecular Cardiovascular Science; Ministry of Education; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Ministry of Health; Beijing Key Laboratory of Cardiovascular Receptors Research; Beijing China
| | - Si Cheng
- China National Clinical Research Center for Neurological Diseases; Tiantan Hospital; The Capital Medical University; Beijing China
| | - Bing Pan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine; School of Basic Medical Sciences; Peking University Health Science Center; Key Laboratory of Molecular Cardiovascular Science; Ministry of Education; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Ministry of Health; Beijing Key Laboratory of Cardiovascular Receptors Research; Beijing China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine; School of Basic Medical Sciences; Peking University Health Science Center; Key Laboratory of Molecular Cardiovascular Science; Ministry of Education; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Ministry of Health; Beijing Key Laboratory of Cardiovascular Receptors Research; Beijing China
- Fujian Medical University Union Hospital; Fuzhou China
| | - Huashan Hong
- Department of Geriatrics; Fujian Medical University Union Hospital; Fuzhou China
| |
Collapse
|
28
|
Dollerup OL, Christensen B, Svart M, Schmidt MS, Sulek K, Ringgaard S, Stødkilde-Jørgensen H, Møller N, Brenner C, Treebak JT, Jessen N. A randomized placebo-controlled clinical trial of nicotinamide riboside in obese men: safety, insulin-sensitivity, and lipid-mobilizing effects. Am J Clin Nutr 2018; 108:343-353. [PMID: 29992272 DOI: 10.1093/ajcn/nqy132] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/17/2018] [Indexed: 12/13/2022] Open
Abstract
Background Animal studies suggest a positive role for nicotinamide riboside (NR) on insulin sensitivity and hepatic steatosis in models of obesity and type 2 diabetes. NR, an NAD+ precursor, is a member of the vitamin B-3 family now available as an over-the-counter supplement. Although data from preclinical trials appear consistent, potential effects and safety need to be evaluated in human clinical trials. Objective The aim of this study was to test the safety of dietary NR supplementation over a 12-wk period and potential to improve insulin sensitivity and other metabolic parameters in obese, insulin-resistant men. Design In an investigator-initiated randomized, placebo-controlled, double-blinded, and parallel-group designed clinical trial, forty healthy, sedentary men with a body mass index (BMI) > 30 kg/m2, age-range 40-70 y were randomly assigned to 12 wk of NR (1000 mg twice daily) or placebo. We determined the effects of NR supplementation on insulin sensitivity by a hyperinsulinemic euglycemic clamp and substrate metabolism by indirect calorimetry and labeled substrates of tritiated glucose and palmitate. Body composition and fat mass distribution were determined by whole-body dual-energy X-ray absorptiometry (DXA) and MRI scans, and measurements of intrahepatic lipid content were obtained by MR spectroscopy. Results Insulin sensitivity, endogenous glucose production, and glucose disposal and oxidation were not improved by NR supplementation. Similarly, NR supplementation had no effect on resting energy expenditure, lipolysis, oxidation of lipids, or body composition. No serious adverse events due to NR supplementation were observed and safety blood tests were normal. Conclusion 12 wk of NR supplementation in doses of 2000 mg/d appears safe, but does not improve insulin sensitivity and whole-body glucose metabolism in obese, insulin-resistant men. This trial was registered at clinicaltrials.gov as NCT02303483.
Collapse
Affiliation(s)
- Ole L Dollerup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Medical Research Laboratory, Department of Clinical Medicine
| | - Britt Christensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Medical Research Laboratory, Department of Clinical Medicine
| | - Mads Svart
- Medical Research Laboratory, Department of Clinical Medicine
| | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Karolina Sulek
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Niels Møller
- Medical Research Laboratory, Department of Clinical Medicine.,Department of Endocrinology
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jessen
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
29
|
Ong AL, Ramasamy TS. Role of Sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res Rev 2018; 43:64-80. [PMID: 29476819 DOI: 10.1016/j.arr.2018.02.004] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/23/2018] [Accepted: 02/16/2018] [Indexed: 12/12/2022]
Abstract
Regulatory role of Sirtuin 1 (SIRT1), one of the most extensively studied members of its kind in histone deacetylase family in governing multiple cellular fates, is predominantly linked to p53 activity. SIRT1 deacetylates p53 in a NAD+-dependent manner to inhibit transcription activity of p53, in turn modulate pathways that are implicated in regulation of tissue homoeostasis and many disease states. In this review, we discuss the role of SIRT1-p53 pathway and its regulatory axis in the cellular events which are implicated in cellular aging, cancer and reprogramming. It is noteworthy that these cellular events share few common regulatory pathways, including SIRT1-p53-LDHA-Myc, miR-34a,-Let7 regulatory network, which forms a positive feedback loop that controls cell cycle, metabolism, proliferation, differentiation, epigenetics and many others. In the context of aging, SIRT1 expression is reduced as a protective mechanism against oncogenesis and for maintenance of tissue homeostasis. Interestingly, its activation in aged cells is evidenced in response to DNA damage to protect the cells from p53-dependent apoptosis or senescence, predispose these cells to neoplastic transformation. Importantly, the dual roles of SIRT1-p53 axis in aging and tumourigenesis, either as tumour suppressor or tumour promoter are determined by SIRT1 localisation and type of cells. Conceptualising the distinct similarity between tumorigenesis and cellular reprogramming, this review provides a perspective discussion on involvement of SIRT1 in improving efficiency in the induction and maintenance of pluripotent state. Further research in understanding the role of SIRT1-p53 pathway and their associated regulators and strategies to manipulate this regulatory axis very likely foster the development of therapeutics and strategies for treating cancer and aging-associated degenerative diseases.
Collapse
|
30
|
Agerholm M, Dall M, Jensen BAH, Prats C, Madsen S, Basse AL, Graae AS, Risis S, Goldenbaum J, Quistorff B, Larsen S, Vienberg SG, Treebak JT. Perturbations of NAD + salvage systems impact mitochondrial function and energy homeostasis in mouse myoblasts and intact skeletal muscle. Am J Physiol Endocrinol Metab 2018; 314:E377-E395. [PMID: 29208611 DOI: 10.1152/ajpendo.00213.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) can be synthesized by nicotinamide phosphoribosyltransferase (NAMPT). We aimed to determine the role of NAMPT in maintaining NAD+ levels, mitochondrial function, and metabolic homeostasis in skeletal muscle cells. We generated stable Nampt knockdown (sh Nampt KD) C2C12 cells using a shRNA lentiviral approach. Moreover, we applied gene electrotransfer to express Cre recombinase in tibialis anterior muscle of floxed Nampt mice. In sh Nampt KD C2C12 myoblasts, Nampt and NAD+ levels were reduced by 70% and 50%, respectively, and maximal respiratory capacity was reduced by 25%. Moreover, anaerobic glycolytic flux increased by 55%, and 2-deoxyglucose uptake increased by 25% in sh Nampt KD cells. Treatment with the NAD+ precursor nicotinamide riboside restored NAD+ levels in sh Nampt cells and increased maximal respiratory capacity by 18% and 32% in control and sh Nampt KD cells, respectively. Expression of Cre recombinase in muscle of floxed Nampt mice reduced NAMPT and NAD+ levels by 38% and 43%, respectively. Glucose uptake increased by 40%, and mitochondrial complex IV respiration was compromised by 20%. Hypoxia-inducible factor (HIF)-1α-regulated genes and histone H3 lysine 9 (H3K9) acetylation, a known sirtuin 6 (SIRT6) target, were increased in shNampt KD cells. Thus, we propose that the shift toward glycolytic metabolism observed, at least in part, is mediated by the SIRT6/HIF1α axis. Our findings suggest that NAMPT plays a key role for maintaining NAD+ levels in skeletal muscle and that NAMPT deficiency compromises oxidative phosphorylation capacity and alters energy homeostasis in this tissue.
Collapse
Affiliation(s)
- Marianne Agerholm
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Morten Dall
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Benjamin A H Jensen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen , Copenhagen , Denmark
| | - Clara Prats
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Søren Madsen
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Astrid L Basse
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Anne-Sofie Graae
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Steve Risis
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Julie Goldenbaum
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Bjørn Quistorff
- Section for Translational Metabolic Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Sara G Vienberg
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Jonas T Treebak
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
31
|
Nielsen KN, Peics J, Ma T, Karavaeva I, Dall M, Chubanava S, Basse AL, Dmytriyeva O, Treebak JT, Gerhart-Hines Z. NAMPT-mediated NAD + biosynthesis is indispensable for adipose tissue plasticity and development of obesity. Mol Metab 2018; 11:178-188. [PMID: 29551635 PMCID: PMC6001355 DOI: 10.1016/j.molmet.2018.02.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/21/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Objective The ability of adipose tissue to expand and contract in response to fluctuations in nutrient availability is essential for the maintenance of whole-body metabolic homeostasis. Given the nutrient scarcity that mammals faced for millions of years, programs involved in this adipose plasticity were likely evolved to be highly efficient in promoting lipid storage. Ironically, this previously advantageous feature may now represent a metabolic liability given the caloric excess of modern society. We speculate that nicotinamide adenine dinucleotide (NAD+) biosynthesis exemplifies this concept. Indeed NAD+/NADH metabolism in fat tissue has been previously linked with obesity, yet whether it plays a causal role in diet-induced adiposity is unknown. Here we investigated how the NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT) supports adipose plasticity and the pathological progression to obesity. Methods We utilized a newly generated Nampt loss-of-function model to investigate the tissue-specific and systemic metabolic consequences of adipose NAD+ deficiency. Energy expenditure, glycemic control, tissue structure, and gene expression were assessed in the contexts of a high dietary fat burden as well as the transition back to normal chow diet. Results Fat-specific Nampt knockout (FANKO) mice were completely resistant to high fat diet (HFD)-induced obesity. This was driven in part by reduced food intake. Furthermore, HFD-fed FANKO mice were unable to undergo healthy expansion of adipose tissue mass, and adipose depots were rendered fibrotic with markedly reduced mitochondrial respiratory capacity. Yet, surprisingly, HFD-fed FANKO mice exhibited improved glucose tolerance compared to control littermates. Removing the HFD burden largely reversed adipose fibrosis and dysfunction in FANKO animals whereas the improved glucose tolerance persisted. Conclusions These findings indicate that adipose NAMPT plays an essential role in handling dietary lipid to modulate fat tissue plasticity, food intake, and systemic glucose homeostasis. Fat-specific Nampt knockout (FANKO) does not alter body composition on chow diet. NAMPT is essential for adipose expansion and weight gain from high dietary fat. Loss of adipose NAD+ decreases food intake and improves glucose tolerance. High fat diet-induced metabolic dysfunction in FANKO mice is reversible.
Collapse
Affiliation(s)
- Karen Nørgaard Nielsen
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Julia Peics
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tao Ma
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Iuliia Karavaeva
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Morten Dall
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sabina Chubanava
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Astrid L Basse
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oksana Dmytriyeva
- Laboratory of Neural Plasticity, Institute of Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark; Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, Copenhagen University Hospital, 2400 Copenhagen, Denmark
| | - Jonas T Treebak
- Section for Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Zachary Gerhart-Hines
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
32
|
LoCoco PM, Risinger AL, Smith HR, Chavera TS, Berg KA, Clarke WP. Pharmacological augmentation of nicotinamide phosphoribosyltransferase (NAMPT) protects against paclitaxel-induced peripheral neuropathy. eLife 2017; 6:e29626. [PMID: 29125463 PMCID: PMC5701795 DOI: 10.7554/elife.29626] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 11/03/2017] [Indexed: 01/03/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) arises from collateral damage to peripheral afferent sensory neurons by anticancer pharmacotherapy, leading to debilitating neuropathic pain. No effective treatment for CIPN exists, short of dose-reduction which worsens cancer prognosis. Here, we report that stimulation of nicotinamide phosphoribosyltransferase (NAMPT) produced robust neuroprotection in an aggressive CIPN model utilizing the frontline anticancer drug, paclitaxel (PTX). Daily treatment of rats with the first-in-class NAMPT stimulator, P7C3-A20, prevented behavioral and histologic indicators of peripheral neuropathy, stimulated tissue NAD recovery, improved general health, and abolished attrition produced by a near maximum-tolerated dose of PTX. Inhibition of NAMPT blocked P7C3-A20-mediated neuroprotection, whereas supplementation with the NAMPT substrate, nicotinamide, potentiated a subthreshold dose of P7C3-A20 to full efficacy. Importantly, P7C3-A20 blocked PTX-induced allodynia in tumored mice without reducing antitumoral efficacy. These findings identify enhancement of NAMPT activity as a promising new therapeutic strategy to protect against anticancer drug-induced peripheral neurotoxicity.
Collapse
Affiliation(s)
- Peter M LoCoco
- Department of PharmacologyUniversity of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - April L Risinger
- Department of PharmacologyUniversity of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Hudson R Smith
- Department of PharmacologyUniversity of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Teresa S Chavera
- Department of PharmacologyUniversity of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - Kelly A Berg
- Department of PharmacologyUniversity of Texas Health Science Center at San AntonioSan AntonioUnited States
| | - William P Clarke
- Department of PharmacologyUniversity of Texas Health Science Center at San AntonioSan AntonioUnited States
| |
Collapse
|
33
|
Kobayashi M, Higami Y. A novel caloric restriction mediator. Aging (Albany NY) 2017; 9:2012-2013. [PMID: 29070730 PMCID: PMC5680547 DOI: 10.18632/aging.101311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/23/2017] [Indexed: 11/25/2022]
Affiliation(s)
- Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
34
|
Homma T, Homma M, Huang Y, Mayurasakorn K, Rodi NM, Hamid AAA, Hurwitz S, Yao T, Adler GK, Pojoga LH, Williams GH, Romero JR. Combined Salt and Caloric Restrictions: Potential Adverse Outcomes. J Am Heart Assoc 2017; 6:e005374. [PMID: 29021272 PMCID: PMC5721821 DOI: 10.1161/jaha.116.005374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/25/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND We hypothesized that caloric restriction (CR) and salt restriction (ResS) would have similar effects on reducing cardiovascular risk markers and that combining CR and ResS would be synergistic in modulating these markers. METHODS AND RESULTS To test our hypothesis, rats were randomized into 2 groups: ad libitum liberal salt diet (ad libitum/high-sodium, 1.6% sodium) or ResS diet (ad libitum/ResS, 0.03% sodium). CR was initiated in half of the rats in each group by reducing caloric intake to 60% while maintaining sodium intake constant (CR/high-sodium, 2.7% sodium or CR/ResS, 0.05% sodium) for 4 weeks. CR in rats on a high-sodium diet improved metabolic parameters, renal transforming growth factor-β and collagen-1α1 and increased plasma adiponectin and renal visfatin and NAD+ protein levels. Although CR produced some beneficial cardiovascular effects (increased sodium excretion and reduced blood pressure), it also was associated with potentially adverse cardiovascular effects. Adrenal zona glomerulosa cell responsiveness and aldosterone levels and activation were inappropriately increased for the volume state of the rodent. Like CR on HS, CR on a ResS diet also produced relative increased zona glomerulosa responsiveness and an increased blood pressure with no improvement in metabolic parameters. CONCLUSIONS These results suggest that combining CR and ResS may decrease the beneficial effects of each alone. Furthermore, CR, regardless of dietary salt intake, inappropriately activates aldosterone production. Thus, caution should be used in combining ResS and CR because the combination may lead to increased cardiovascular risk.
Collapse
Affiliation(s)
- Tsuyoshi Homma
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Mika Homma
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Yuefei Huang
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Korapat Mayurasakorn
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Nurul Mahamad Rodi
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | | | - Shelley Hurwitz
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Tham Yao
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Gail K Adler
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Jose R Romero
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
35
|
Abstract
Vascular repair plays important roles in postischemic remodeling and rehabilitation in cardiovascular and cerebrovascular disease, such as stroke and myocardial infarction. Nicotinamide adenine dinucleotide (NAD), a well-known coenzyme involved in electron transport chain for generation of adenosine triphosphate, has emerged as an important controller regulating various biological signaling pathways. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme for NAD biosynthesis in mammals. NAMPT may also act in a nonenzymatic manner, presumably mediated by unknown receptor(s). Rapidly accumulating data in the past decade show that NAMPT and NAMPT-controlled NAD metabolism regulate fundamental biological functions in endothelial cells, vascular smooth muscle cells, and endothelial progenitor cells. The NAD-consuming proteins, including sirtuins, poly-ADP-ribose polymerases (PARPs), and CD38, may contribute to the regulatory effects of NAMPT-NAD axis in these cells and vascular repair. This review discusses the current data regarding NAMPT and NAMPT-controlled NAD metabolism in vascular repair and the clinical potential translational application of NAMPT-related products in treatment of cardiovascular and cerebrovascular disease.
Collapse
|
36
|
Jenwitheesuk A, Park S, Wongchitrat P, Tocharus J, Mukda S, Shimokawa I, Govitrapong P. Comparing the Effects of Melatonin with Caloric Restriction in the Hippocampus of Aging Mice: Involvement of Sirtuin1 and the FOXOs Pathway. Neurochem Res 2017; 43:153-161. [PMID: 28770437 DOI: 10.1007/s11064-017-2369-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022]
Abstract
It has been suggested that age-related neurodegeneration might be associated with neuropeptide Y (NPY); sirtuin1 (SIRT1) and forkhead box transcription factors O subfamily (FOXOs) pathways. Melatonin, a hormone mainly secreted by the pineal gland, is another anti-aging agent associated with the SIRT1-FOXOs pathway. This study aimed to compare the effects of melatonin (Mel) and caloric restriction (CR) on the expression of Sirt1, FoxO1, FoxO3a and FOXOs target genes in the aging mouse hippocampus. Neuropeptide Y-knockout (NpyKO) and wild-type (WT) male mice aged 19 months were previously treated either with food ad libitum or CR for 16 months. WT old animals were divided into four groups: control, CR, Mel and CR+Mel treated groups. The Mel and CR+Mel were treated with melatonin 10 mg/kg, daily, subcutaneously for 7 consecutive days. Mel treatment upregulated the mRNA expression of Sirt1, FOXOs (FoxO1 and FoxO3a) target genes that regulated the cell cycle [e.g., cyclin-dependent kinase inhibitor 1B (p27)], Wingless and INT-1 (Wnt1) and inducible signaling pathway protein 1 (Wisp1) in the aged mouse hippocampus. CR treatment also showed the similar actions. However, the mRNA expression of Sirt1, FoxO1, FoxO3a, p27 or Wisp1 did not alter in the CR+Mel group when compared with CR or Mel group. Melatonin could not produce any additive effect on the CR treatment group, suggesting that both treatments mimicked the effect, possibly via the same pathway. NPY which mediates physiological adaptations to energy deficits is an essential link between CR and longevity in mice. In order to focus on the role of Npy in mediating the effects of melatonin, the gene expression between NpyKO and WT male mice were compared. Our data showed that, in the absence of Npy, melatonin could not mediate effects on those gene expressions, suggesting that Npy was required for melatonin to mediate the effect, possibly, on life extension.
Collapse
Affiliation(s)
- Anorut Jenwitheesuk
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Seongjoon Park
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan
| | - Prapimpun Wongchitrat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom, 73170, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand
| | - Isao Shimokawa
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan.
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand. .,Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol University, Salaya, Thailand. .,Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Kamphaeng Phet 6 Road, Lak Si, Bangkok, 10210, Thailand.
| |
Collapse
|
37
|
Chang M, Qiao L, Li B, Wang J, Zhang G, Shi W, Liu Z, Gu N, Di Z, Wang X, Tian Y. Suppression of SIRT6 by miR-33a facilitates tumor growth of glioma through apoptosis and oxidative stress resistance. Oncol Rep 2017; 38:1251-1258. [DOI: 10.3892/or.2017.5780] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 06/23/2017] [Indexed: 01/17/2023] Open
|
38
|
Elhassan YS, Philp AA, Lavery GG. Targeting NAD+ in Metabolic Disease: New Insights Into an Old Molecule. J Endocr Soc 2017; 1:816-835. [PMID: 29264533 PMCID: PMC5686634 DOI: 10.1210/js.2017-00092] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/10/2017] [Indexed: 02/06/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an established cofactor for enzymes serving cellular metabolic reactions. More recent research identified NAD+ as a signaling molecule and substrate for sirtuins and poly-adenosine 5'-diphosphate polymerases; enzymes that regulate protein deacetylation and DNA repair, and translate changes in energy status into metabolic adaptations. Deranged NAD+ homeostasis and concurrent alterations in mitochondrial function are intrinsic in metabolic disorders, such as type 2 diabetes, nonalcoholic fatty liver, and age-related diseases. Contemporary NAD+ precursors show promise as nutraceuticals to restore target tissue NAD+ and have demonstrated the ability to improve mitochondrial function and sirtuin-dependent signaling. This review discusses the accumulating evidence for targeting NAD+ metabolism in metabolic disease, maps the different strategies for NAD+ boosting, and addresses the challenges and open questions in the field. The health potential of targeting NAD+ homeostasis will inform clinical study design to identify nutraceutical approaches for combating metabolic disease and the unwanted effects of aging.
Collapse
Affiliation(s)
- Yasir S. Elhassan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, United Kingdom
| | - Andrew A. Philp
- MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Gareth G. Lavery
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
39
|
Fujii N, Narita T, Okita N, Kobayashi M, Furuta Y, Chujo Y, Sakai M, Yamada A, Takeda K, Konishi T, Sudo Y, Shimokawa I, Higami Y. Sterol regulatory element-binding protein-1c orchestrates metabolic remodeling of white adipose tissue by caloric restriction. Aging Cell 2017; 16:508-517. [PMID: 28256090 PMCID: PMC5418191 DOI: 10.1111/acel.12576] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2017] [Indexed: 12/31/2022] Open
Abstract
Caloric restriction (CR) can delay onset of several age‐related pathophysiologies and extend lifespan in various species, including rodents. CR also induces metabolic remodeling involved in activation of lipid metabolism, enhancement of mitochondrial biogenesis, and reduction of oxidative stress in white adipose tissue (WAT). In studies using genetically modified mice with extended lifespans, WAT characteristics influenced mammalian lifespans. However, molecular mechanisms underlying CR‐associated metabolic remodeling of WAT remain unclear. Sterol regulatory element‐binding protein‐1c (Srebp‐1c), a master transcription factor of fatty acid (FA) biosynthesis, is responsible for the pathogenesis of fatty liver (steatosis). Our study showed that, under CR conditions, Srebp‐1c enhanced mitochondrial biogenesis via increased expression of peroxisome proliferator‐activated receptor gamma coactivator‐1α (Pgc‐1α) and upregulated expression of proteins involved in FA biosynthesis within WAT. However, via Srebp‐1c, most of these CR‐associated metabolic alterations were not observed in other tissues, including the liver. Moreover, our data indicated that Srebp‐1c may be an important factor both for CR‐associated suppression of oxidative stress, through increased synthesis of glutathione in WAT, and for the prolongevity action of CR. Our results strongly suggested that Srebp‐1c, the primary FA biosynthesis‐promoting transcriptional factor implicated in fatty liver disease, is also the food shortage‐responsive factor in WAT. This indicated that Srebp‐1c is a key regulator of metabolic remodeling leading to the beneficial effects of CR.
Collapse
Affiliation(s)
- Namiki Fujii
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Takumi Narita
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Naoyuki Okita
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Department of Internal Medicine Research; Sasaki Institute; Sasaki Foundation; 2-2 Kandasurugadai Chiyoda-ku, Tokyo 101-0062 Japan
| | - Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Yurika Furuta
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Yoshikazu Chujo
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Masahiro Sakai
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Atsushi Yamada
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Kanae Takeda
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Tomokazu Konishi
- Faculty of Bioresource Sciences; Akita Prefectural University; Shimoshinjo Nakano, Akita 010-0195 Japan
| | - Yuka Sudo
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| | - Isao Shimokawa
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Department of Pathology; Nagasaki University Graduate School of Biomedical Sciences; 1-12-4 Sakamoto Nagasaki 852-8523 Japan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease; Faculty of Pharmaceutical Sciences; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
- Translational Research Center, Research Institute of Science and Technology; Tokyo University of Science; 2641 Yamazaki Noda, Chiba 278-8510 Japan
| |
Collapse
|
40
|
Yamaguchi S, Yoshino J. Adipose tissue NAD + biology in obesity and insulin resistance: From mechanism to therapy. Bioessays 2017; 39. [PMID: 28295415 DOI: 10.1002/bies.201600227] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+ ) biosynthetic pathway, mediated by nicotinamide phosphoribosyltransferase (NAMPT), a key NAD+ biosynthetic enzyme, plays a pivotal role in controlling many biological processes, such as metabolism, circadian rhythm, inflammation, and aging. Over the past decade, NAMPT-mediated NAD+ biosynthesis, together with its key downstream mediator, namely the NAD+ -dependent protein deacetylase SIRT1, has been demonstrated to regulate glucose and lipid metabolism in a tissue-dependent manner. These discoveries have provided novel mechanistic and therapeutic insights into obesity and its metabolic complications, such as insulin resistance, an important risk factor for developing type 2 diabetes and cardiovascular disease. This review will focus on the importance of adipose tissue NAMPT-mediated NAD+ biosynthesis and SIRT1 in the pathophysiology of obesity and insulin resistance. We will also critically explore translational and clinical aspects of adipose tissue NAD+ biology.
Collapse
Affiliation(s)
- Shintaro Yamaguchi
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
41
|
Jokinen R, Pirnes-Karhu S, Pietiläinen KH, Pirinen E. Adipose tissue NAD +-homeostasis, sirtuins and poly(ADP-ribose) polymerases -important players in mitochondrial metabolism and metabolic health. Redox Biol 2017; 12:246-263. [PMID: 28279944 PMCID: PMC5343002 DOI: 10.1016/j.redox.2017.02.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/13/2017] [Indexed: 12/14/2022] Open
Abstract
Obesity, a chronic state of energy overload, is characterized by adipose tissue dysfunction that is considered to be the major driver for obesity associated metabolic complications. The reasons for adipose tissue dysfunction are incompletely understood, but one potential contributing factor is adipose tissue mitochondrial dysfunction. Derangements of adipose tissue mitochondrial biogenesis and pathways associate with obesity and metabolic diseases. Mitochondria are central organelles in energy metabolism through their role in energy derivation through catabolic oxidative reactions. The mitochondrial processes are dependent on the proper NAD+/NADH redox balance and NAD+ is essential for reactions catalyzed by the key regulators of mitochondrial metabolism, sirtuins (SIRTs) and poly(ADP-ribose) polymerases (PARPs). Notably, obesity is associated with disturbed adipose tissue NAD+ homeostasis and the balance of SIRT and PARP activities. In this review we aim to summarize existing literature on the maintenance of intracellular NAD+ pools and the function of SIRTs and PARPs in adipose tissue during normal and obese conditions, with the purpose of comprehending their potential role in mitochondrial derangements and obesity associated metabolic complications. Understanding the molecular mechanisms that are the root cause of the adipose tissue mitochondrial derangements is crucial for developing new effective strategies to reverse obesity associated metabolic complications.
Collapse
Affiliation(s)
- Riikka Jokinen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, Biomedicum Helsinki, University of Helsinki, Biomedicum Helsinki, Helsinki, Finland
| | - Sini Pirnes-Karhu
- Molecular Neurology, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, Biomedicum Helsinki, University of Helsinki, Biomedicum Helsinki, Helsinki, Finland; Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland; FIMM, Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Eija Pirinen
- Molecular Neurology, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
42
|
Ma C, Pi C, Yang Y, Lin L, Shi Y, Li Y, Li Y, He X. Nampt Expression Decreases Age-Related Senescence in Rat Bone Marrow Mesenchymal Stem Cells by Targeting Sirt1. PLoS One 2017; 12:e0170930. [PMID: 28125705 PMCID: PMC5268649 DOI: 10.1371/journal.pone.0170930] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/12/2017] [Indexed: 02/07/2023] Open
Abstract
Senescence restricts the development of applications involving mesenchymal stem cells (MSCs) in research fields, such as tissue engineering, and stem cell therapeutic strategies. Understanding the mechanisms underlying natural aging processes may contribute to the development of novel approaches to preventing age-related diseases or slowing individual aging processes. Nampt is a rate-limiting NAD biosynthetic enzyme that plays critical roles in energy metabolism, cell senescence and maintaining life spans. However, it remains unknown whether Nampt influences stem cell senescence. In this study, the function of Nampt was investigated using a rat model of natural aging. Our data show that Nampt expression was significantly lower in MSCs obtained from aged rats than in those obtained from young rats during physiological aging. Reducing the level of Nampt in aged MSCs resulted in lower intracellular concentrations of NAD+ and downregulated Sirt1 expression and activity. After the Nampt inhibitor FK866 was added, young MSCs were induced to become aged cells. The enhanced senescence was correlated with NAD+ depletion and Sirt1 activity attenuation. In addition, Nampt overexpression attenuated cell senescence in aged MSCs. Our findings provide a new explanation for the mechanisms underlying stem cell senescence and a novel target for delaying stem cell senescence and preventing and treating age-related diseases.
Collapse
Affiliation(s)
- Cao Ma
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chenchen Pi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yue Yang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lin Lin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yingai Shi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yan Li
- Center for Advanced Reconstruction of Extremities (C.A.R.E.), Sahlgrenska University Hospital, Mölndal, Sweden
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
43
|
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronizes these with daily cycles, feeding patterns also regulates circadian clocks. The clock genes and adipocytokines show circadian rhythmicity. Dysfunction of these genes are involved in the alteration of these adipokines during the development of obesity. Food availability promotes the stimuli associated with food intake which is a circadian oscillator outside of the suprachiasmatic nucleus (SCN). Its circadian rhythm is arranged with the predictable daily mealtimes. Food anticipatory activity is mediated by a self-sustained circadian timing and its principal component is food entrained oscillator. However, the hypothalamus has a crucial role in the regulation of energy balance rather than food intake. Fatty acids or their metabolites can modulate neuronal activity by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. The timing of three-meal schedules indicates close association with the plasma levels of insulin and preceding food availability. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition can lead to uncoupling of peripheral clocks from the central pacemaker and to the development of metabolic disorders. Metabolic dysfunction is associated with circadian disturbances at both central and peripheral levels and, eventual disruption of circadian clock functioning can lead to obesity. While CLOCK expression levels are increased with high fat diet-induced obesity, peroxisome proliferator-activated receptor (PPAR) alpha increases the transcriptional level of brain and muscle ARNT-like 1 (BMAL1) in obese subjects. Consequently, disruption of clock genes results in dyslipidemia, insulin resistance and obesity. Modifying the time of feeding alone can greatly affect body weight. Changes in the circadian clock are associated with temporal alterations in feeding behavior and increased weight gain. Thus, shift work is associated with increased risk for obesity, diabetes and cardio-vascular diseases as a result of unusual eating time and disruption of circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
44
|
Inhibitor of Nicotinamide Phosphoribosyltransferase Sensitizes Glioblastoma Cells to Temozolomide via Activating ROS/JNK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1450843. [PMID: 28097126 PMCID: PMC5206411 DOI: 10.1155/2016/1450843] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022]
Abstract
Overcoming temozolomide (TMZ) resistance is a great challenge in glioblastoma (GBM) treatment. Nicotinamide phosphoribosyltransferase (NAMPT) is a rate-limiting enzyme in the biosynthesis of nicotinamide adenine dinucleotide and has a crucial role in cancer cell metabolism. In this study, we investigated whether FK866 and CHS828, two specific NAMPT inhibitors, could sensitize GBM cells to TMZ. Low doses of FK866 and CHS828 (5 nM and 10 nM, resp.) alone did not significantly decrease cell viability in U251-MG and T98 GBM cells. However, they significantly increased the antitumor action of TMZ in these cells. In U251-MG cells, administration of NAMPT inhibitors increased the TMZ (100 μM)-induced apoptosis and LDH release from GBM cells. NAMPT inhibitors remarkably enhanced the activities of caspase-1, caspase-3, and caspase-9. Moreover, NAMPT inhibitors increased reactive oxygen species (ROS) production and superoxide anion level but reduced the SOD activity and total antioxidative capacity in GBM cells. Treatment of NAMPT inhibitors increased phosphorylation of c-Jun and JNK. Administration of JNK inhibitor SP600125 or ROS scavenger tocopherol with TMZ and NAMPT inhibitors substantially attenuated the sensitization of NAMPT inhibitor on TMZ antitumor action. Our data indicate a potential value of NAMPT inhibitors in combined use with TMZ for GBM treatment.
Collapse
|
45
|
Yang Y, Sauve AA. NAD(+) metabolism: Bioenergetics, signaling and manipulation for therapy. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1864:1787-1800. [PMID: 27374990 PMCID: PMC5521000 DOI: 10.1016/j.bbapap.2016.06.014] [Citation(s) in RCA: 319] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/30/2016] [Accepted: 06/28/2016] [Indexed: 12/19/2022]
Abstract
We survey the historical development of scientific knowledge surrounding Vitamin B3, and describe the active metabolite forms of Vitamin B3, the pyridine dinucleotides NAD+ and NADP+ which are essential to cellular processes of energy metabolism, cell protection and biosynthesis. The study of NAD+ has become reinvigorated by new understandings that dynamics within NAD+ metabolism trigger major signaling processes coupled to effectors (sirtuins, PARPs, and CD38) that reprogram cellular metabolism using NAD+ as an effector substrate. Cellular adaptations include stimulation of mitochondrial biogenesis, a process fundamental to adjusting cellular and tissue physiology to reduced nutrient availability and/or increased energy demand. Several mammalian metabolic pathways converge to NAD+, including tryptophan-derived de novo pathways, nicotinamide salvage pathways, nicotinic acid salvage and nucleoside salvage pathways incorporating nicotinamide riboside and nicotinic acid riboside. Key discoveries highlight a therapeutic potential for targeting NAD+ biosynthetic pathways for treatment of human diseases. A recent emergence of understanding that NAD+ homeostasis is vulnerable to aging and disease processes has stimulated testing to determine if replenishment or augmentation of cellular or tissue NAD+ can have ameliorative effects on aging or disease phenotypes. This experimental approach has provided several proofs of concept successes demonstrating that replenishment or augmentation of NAD+ concentrations can provide ameliorative or curative benefits. Thus NAD+ metabolic pathways can provide key biomarkers and parameters for assessing and modulating organism health.
Collapse
Affiliation(s)
- Yue Yang
- Department of Pharmacology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
46
|
Sparks LM, Redman LM, Conley KE, Harper ME, Hodges A, Eroshkin A, Costford SR, Gabriel ME, Yi F, Shook C, Cornnell HH, Ravussin E, Smith SR. Differences in Mitochondrial Coupling Reveal a Novel Signature of Mitohormesis in Muscle of Healthy Individuals. J Clin Endocrinol Metab 2016; 101:4994-5003. [PMID: 27710240 PMCID: PMC5155692 DOI: 10.1210/jc.2016-2742] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
CONTEXT Reduced mitochondrial coupling (ATP/O2 [P/O]) is associated with sedentariness and insulin resistance. Interpreting the physiological relevance of P/O measured in vitro is challenging. OBJECTIVE To evaluate muscle mitochondrial function and associated transcriptional profiles in nonobese healthy individuals distinguished by their in vivo P/O. DESIGN Individuals from an ancillary study of Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy phase 2 were assessed at baseline. SETTING The study was performed at Pennington Biomedical Research Center. PARTICIPANTS Forty-seven (18 males, 26-50 y of age) sedentary, healthy nonobese individuals were divided into 2 groups based on their in vivo P/O. INTERVENTION None. Main Outcome(s): Body composition by dual-energy x-ray absorptiometry, in vivo mitochondrial function (P/O and maximal ATP synthetic capacity) by 31P-magnetic resonance spectroscopy and optical spectroscopy were measured. A muscle biopsy was performed to measure fiber type, transcriptional profiling (microarray), and protein expressions. RESULTS No differences in body composition, peak aerobic capacity, type I fiber content, or mitochondrial DNA copy number were observed between the 2 groups. Compared with the uncoupled group (lower P/O), the coupled group (higher P/O) had higher rates of maximal ATP synthetic capacity (maximal ATP synthetic capacity, P < .01). Transcriptomics analyses revealed higher expressions of genes involved in mitochondrial remodeling and the oxidative stress response in the coupled group. A trend for higher mitonuclear protein imbalance (P = .06) and an elevated mitochondrial unfolded protein response (heat shock protein 60 protein; P = .004) were also identified in the coupled group. CONCLUSIONS Higher muscle mitochondrial coupling is accompanied by an overall elevation in mitochondrial function, a novel transcriptional signature of oxidative stress and mitochondrial remodeling and indications of an mitochondrial unfolded protein response.
Collapse
Affiliation(s)
- Lauren M Sparks
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Leanne M Redman
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Kevin E Conley
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Mary-Ellen Harper
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Andrew Hodges
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Alexey Eroshkin
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Sheila R Costford
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Meghan E Gabriel
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Fanchao Yi
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Cherie Shook
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Heather H Cornnell
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Eric Ravussin
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| |
Collapse
|
47
|
Imai SI, Guarente L. It takes two to tango: NAD + and sirtuins in aging/longevity control. NPJ Aging Mech Dis 2016; 2:16017. [PMID: 28721271 PMCID: PMC5514996 DOI: 10.1038/npjamd.2016.17] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 12/14/2022] Open
Abstract
The coupling of nicotinamide adenine dinucleotide (NAD+) breakdown and protein deacylation is a unique feature of the family of proteins called ‘sirtuins.’ This intimate connection between NAD+ and sirtuins has an ancient origin and provides a mechanistic foundation that translates the regulation of energy metabolism into aging and longevity control in diverse organisms. Although the field of sirtuin research went through intensive controversies, an increasing number of recent studies have put those controversies to rest and fully established the significance of sirtuins as an evolutionarily conserved aging/longevity regulator. The tight connection between NAD+ and sirtuins is regulated at several different levels, adding further complexity to their coordination in metabolic and aging/longevity control. Interestingly, it has been demonstrated that NAD+ availability decreases over age, reducing sirtuin activities and affecting the communication between the nucleus and mitochondria at a cellular level and also between the hypothalamus and adipose tissue at a systemic level. These dynamic cellular and systemic processes likely contribute to the development of age-associated functional decline and the pathogenesis of diseases of aging. To mitigate these age-associated problems, supplementation of key NAD+ intermediates is currently drawing significant attention. In this review article, we will summarize these important aspects of the intimate connection between NAD+ and sirtuins in aging/longevity control.
Collapse
Affiliation(s)
- Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Leonard Guarente
- Department of Biology and Glenn Laboratories for the Science of Aging, Massachusetts Institute of Technology, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
48
|
Stromsdorfer KL, Yamaguchi S, Yoon MJ, Moseley AC, Franczyk MP, Kelly SC, Qi N, Imai SI, Yoshino J. NAMPT-Mediated NAD(+) Biosynthesis in Adipocytes Regulates Adipose Tissue Function and Multi-organ Insulin Sensitivity in Mice. Cell Rep 2016; 16:1851-60. [PMID: 27498863 PMCID: PMC5094180 DOI: 10.1016/j.celrep.2016.07.027] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 06/20/2016] [Accepted: 07/13/2016] [Indexed: 01/14/2023] Open
Abstract
Obesity is associated with adipose tissue dysfunction and multi-organ insulin resistance. However, the mechanisms of such obesity-associated systemic metabolic complications are not clear. Here, we characterized mice with adipocyte-specific deletion of nicotinamide phosphoribosyltransferase (NAMPT), a rate-limiting NAD(+) biosynthetic enzyme known to decrease in adipose tissue of obese and aged rodents and people. We found that adipocyte-specific Nampt knockout mice had severe insulin resistance in adipose tissue, liver, and skeletal muscle and adipose tissue dysfunction, manifested by increased plasma free fatty acid concentrations and decreased plasma concentrations of a major insulin-sensitizing adipokine, adiponectin. Loss of Nampt increased phosphorylation of CDK5 and PPARγ (serine-273) and decreased gene expression of obesity-linked phosphorylated PPARγ targets in adipose tissue. These deleterious alterations were normalized by administering rosiglitazone or a key NAD(+) intermediate, nicotinamide mononucleotide (NMN). Collectively, our results provide important mechanistic and therapeutic insights into obesity-associated systemic metabolic derangements, particularly multi-organ insulin resistance.
Collapse
Affiliation(s)
- Kelly L Stromsdorfer
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shintaro Yamaguchi
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Myeong Jin Yoon
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anna C Moseley
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael P Franczyk
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shannon C Kelly
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nathan Qi
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun Yoshino
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
49
|
Wang P, Yang X, Zhang Z, Song J, Guan YF, Zou DJ, Miao CY. Depletion of NAD pool contributes to impairment of endothelial progenitor cell mobilization in diabetes. Metabolism 2016; 65:852-62. [PMID: 27173464 DOI: 10.1016/j.metabol.2016.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/07/2016] [Accepted: 03/09/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The impaired mobilization of endothelial progenitor cells (EPCs) from bone marrow (BM) critically contributes to the diabetes-associated vascular complications. Here, we investigated the relationship between the nicotinamide phosphoribosyltransferase (NAMPT)-controlled nicotinamide adenine dinucleotide (NAD) metabolism and the impaired mobilization of BM-derived EPCs in diabetic condition. METHODS The NAMPT-NAD pool in BM and BM-derived EPCs in wild-type (WT) and diabetic db/db mice was determined. Nicotinamide, a natural substrate for NAD biosynthesis, was administrated for 2weeks in db/db mice to examine the influence of enhancing NAD pool on BM and blood EPCs number. The modulations of stromal cell-derived factor-1α (SDF-1α) and endothelial nitric oxide synthase (eNOS) protein in BM were measured using immunoblotting. The EPCs intracellular NAMPT level and NAD concentration, as well as the blood EPCs number, were compared between 9 healthy people and 16 patients with type 2 diabetes mellitus (T2DM). The T2DM patients were treated with nicotinamide for two weeks and then the blood EPCs number was determined. Moreover, the association between blood EPCs numbers and EPCs intracellular NAD(+)/NAMPT protein levels in 21 healthy individuals was determined. RESULTS We found that NAD concentration and NAMPT expression in BM and BM-derived EPCs of db/db mice were significantly lower than those in WT mice BM. Enhancing NAD pool not only increased the EPCs intracellular NAD concentration and blood EPCs number, but also improved post-ischemic wound healing and blood reperfusion in db/db mice with hind-limb ischemia model. Enhancing NAD pool rescued the impaired modulations of stromal cell-derived factor-1α (SDF-1α) and endothelial nitric oxide synthase (eNOS) protein levels in db/db mice BM upon hind-limb ischemia. In addition, enhancing NAD pool significantly inhibited PARP and caspase-3 activates in db/db mice BM. The intracellular NAMPT-NAD pool was positively associated with blood EPCs number in healthy individuals. At last, we found that the EPC intracellular NAMPT and NAD(+) levels were reduced in T2DM patients and enhancing NAD pool elevated the circulating blood EPCs number in T2DM patients. CONCLUSION Our results indicate that the depletion of NAD pool may contribute to the impairment of EPCs mobilization in diabetic condition, and imply the potential therapeutic value of nicotinamide in the prevention and treatment for cardiovascular complications of diabetes.
Collapse
Affiliation(s)
- Pei Wang
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China; Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Xi Yang
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zheng Zhang
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jie Song
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Yun-Feng Guan
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Da-Jin Zou
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University, Shanghai, China.
| |
Collapse
|
50
|
Zheng SL, Li ZY, Song J, Liu JM, Miao CY. Metrnl: a secreted protein with new emerging functions. Acta Pharmacol Sin 2016; 37:571-9. [PMID: 27063217 PMCID: PMC4857552 DOI: 10.1038/aps.2016.9] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/24/2016] [Indexed: 12/15/2022]
Abstract
Secreted proteins play critical roles in physiological and pathological processes and can be used as biomarkers and therapies for aging and disease. Metrnl is a novel secreted protein homologous to the neurotrophin Metrn. But this protein, unlike Metrn that is mainly expressed in the brain, shows a relatively wider distribution in the body with high levels of expression in white adipose tissue and barrier tissues. This protein plays important roles in neural development, white adipose browning and insulin sensitization. Based on its expression and distinct functions, this protein is also called Cometin, Subfatin and Interleukin 39, which refer to its neurotrophic effect, adipokine function and the possible action as a cytokine, respectively. The spectrum of Metrnl functions remains to be determined, and the mechanisms of Metrnl action need to be elucidated. In this review, we focus on the discovery, structural characteristics, expression pattern and physiological functions of Metrnl, which will assist in developing this protein as a new therapeutic target or agent.
Collapse
Affiliation(s)
- Si-li Zheng
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Zhi-yong Li
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Jie Song
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Jian-min Liu
- Stroke Center & Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chao-yu Miao
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100069, China
- E-mail
| |
Collapse
|