1
|
Pańczyszyn A, Boniewska-Bernacka E, Wertel I, Sadakierska-Chudy A, Goc A. Telomeres and SIRT1 as Biomarkers of Gamete Oxidative Stress, Fertility, and Potential IVF Outcome. Int J Mol Sci 2024; 25:8652. [PMID: 39201341 PMCID: PMC11354255 DOI: 10.3390/ijms25168652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
The number of infertile couples undergoing in vitro fertilisation (IVF) has increased significantly. The efficacy of this procedure is contingent upon a multitude of factors, including gamete quality. One factor influencing gamete quality is oxidative stress, which leads to telomere damage and accelerates cellular ageing. Identifying new biomarkers that can predict the success of assisted reproduction techniques is a current relevant area of research. In this review, we discuss the potential role of SIRT1, a protein known to protect against oxidative stress and telomeres, which are responsible for genome stability, as biomarkers of gamete quality and assisted reproduction technique outcomes.
Collapse
Affiliation(s)
- Anna Pańczyszyn
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052 Opole, Poland; (E.B.-B.); (A.G.)
| | - Ewa Boniewska-Bernacka
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052 Opole, Poland; (E.B.-B.); (A.G.)
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland;
| | - Anna Sadakierska-Chudy
- Department of Genetics, Faculty of Medicine and Health Sciences, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705 Krakow, Poland;
| | - Anna Goc
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052 Opole, Poland; (E.B.-B.); (A.G.)
| |
Collapse
|
2
|
Yu X, Li S. Specific regulation of epigenome landscape by metabolic enzymes and metabolites. Biol Rev Camb Philos Soc 2024; 99:878-900. [PMID: 38174803 DOI: 10.1111/brv.13049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024]
Abstract
Metabolism includes anabolism and catabolism, which play an essential role in many biological processes. Chromatin modifications are post-translational modifications of histones and nucleic acids that play important roles in regulating chromatin-associated processes such as gene transcription. There is a tight connection between metabolism and chromatin modifications. Many metabolic enzymes and metabolites coordinate cellular activities with alterations in nutrient availability by regulating gene expression through epigenetic mechanisms such as DNA methylation and histone modifications. The dysregulation of gene expression by metabolism and epigenetic modifications may lead to diseases such as diabetes and cancer. Recent studies reveal that metabolic enzymes and metabolites specifically regulate chromatin modifications, including modification types, modification residues and chromatin regions. This specific regulation has been implicated in the development of human diseases, yet the underlying mechanisms are only beginning to be uncovered. In this review, we summarise recent studies of the molecular mechanisms underlying the metabolic regulation of histone and DNA modifications and discuss how they contribute to pathogenesis. We also describe recent developments in technologies used to address the key questions in this field. We hope this will inspire further in-depth investigations of the specific regulatory mechanisms involved, and most importantly will shed lights on the development of more effective disease therapies.
Collapse
Affiliation(s)
- Xilan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Shanshan Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| |
Collapse
|
3
|
Rogina B, Tissenbaum HA. SIRT1, resveratrol and aging. Front Genet 2024; 15:1393181. [PMID: 38784035 PMCID: PMC11112063 DOI: 10.3389/fgene.2024.1393181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Aging is linked to a time-associated decline in both cellular function and repair capacity leading to malfunction on an organismal level, increased frailty, higher incidence of diseases, and death. As the population grows older, there is a need to reveal mechanisms associated with aging that could spearhead treatments to postpone the onset of age-associated decline, extend both healthspan and lifespan. One possibility is targeting the sirtuin SIRT1, the founding member of the sirtuin family, a highly conserved family of histone deacetylases that have been linked to metabolism, stress response, protein synthesis, genomic instability, neurodegeneration, DNA damage repair, and inflammation. Importantly, sirtuins have also been implicated to promote health and lifespan extension, while their dysregulation has been linked to cancer, neurological processes, and heart disorders. SIRT1 is one of seven members of sirtuin family; each requiring nicotinamide adenine dinucleotide (NAD+) as co-substrate for their catalytic activity. Overexpression of yeast, worm, fly, and mice SIRT1 homologs extend lifespan in each animal, respectively. Moreover, lifespan extension due to calorie restriction are associated with increased sirtuin activity. These findings led to the search for a calorie restriction mimetic, which revealed the compound resveratrol; (3, 5, 4'-trihydroxy-trans-stilbene) belonging to the stilbenoids group of polyphenols. Following this finding, resveratrol and other sirtuin-activating compounds have been extensively studied for their ability to affect health and lifespan in a variety of species, including humans via clinical studies.
Collapse
Affiliation(s)
- Blanka Rogina
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
- Institute for Systems Genomics, Farmington, CT, United States
| | - Heidi A. Tissenbaum
- Department of Molecular, Cell and Cancer Biology UMass Chan Medical School, Worcester, MA, United States
| |
Collapse
|
4
|
Radulescu D, Mihai FD, Trasca MET, Caluianu EI, Calafeteanu CDM, Radulescu PM, Mercut R, Ciupeanu-Calugaru ED, Marinescu GA, Siloşi CA, Nistor CCE, Danoiu S. Oxidative Stress in Military Missions-Impact and Management Strategies: A Narrative Analysis. Life (Basel) 2024; 14:567. [PMID: 38792589 PMCID: PMC11121804 DOI: 10.3390/life14050567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
This narrative review comprehensively examines the impact of oxidative stress on military personnel, highlighting the crucial role of physical exercise and tailored diets, particularly the ketogenic diet, in minimizing this stress. Through a meticulous analysis of the recent literature, the study emphasizes how regular physical exercise not only enhances cardiovascular, cognitive, and musculoskeletal health but is also essential in neutralizing the effects of oxidative stress, thereby improving endurance and performance during long-term missions. Furthermore, the implementation of the ketogenic diet provides an efficient and consistent energy source through ketone bodies, tailored to the specific energy requirements of military activities, and significantly contributes to the reduction in reactive oxygen species production, thus protecting against cellular deterioration under extreme stress. The study also underlines the importance of integrating advanced technologies, such as wearable devices and smart sensors that allow for the precise and real-time monitoring of oxidative stress and physiological responses, thus facilitating the customization of training and nutritional regimes. Observations from this review emphasize significant variability among individuals in responses to oxidative stress, highlighting the need for a personalized approach in formulating intervention strategies. It is crucial to develop and implement well-monitored, personalized supplementation protocols to ensure that each member of the military personnel receives a regimen tailored to their specific needs, thereby maximizing the effectiveness of measures to combat oxidative stress. This analysis makes a valuable contribution to the specialized literature, proposing a detailed framework for addressing oxidative stress in the armed forces and opening new directions for future research with the aim of optimizing clinical practices and improving the health and performance of military personnel under stress and specific challenges of the military field.
Collapse
Affiliation(s)
- Dumitru Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Florina-Diana Mihai
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | - Major Emil-Tiberius Trasca
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Elena-Irina Caluianu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Captain Dan Marian Calafeteanu
- Department of Ortopedics, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania;
| | - Patricia-Mihaela Radulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Razvan Mercut
- Department of Plastic and Reconstructive Surgery, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | | | - Georgiana-Andreea Marinescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’ Craiova, 200749 Craiova, Romania; (D.R.); (E.-I.C.); (P.-M.R.); (G.-A.M.)
| | - Cristian-Adrian Siloşi
- Doctoral School, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | | | - Suzana Danoiu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
5
|
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: 8.0] [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
|
6
|
Bohnen JLB, Wigstrom TP, Griggs AM, Roytman S, Paalanen RR, Andrews HA, Bohnen NI, Franklin JJH, McInnis MG. Ketogenic-Mimicking Diet as a Therapeutic Modality for Bipolar Disorder: Biomechanistic Rationale and Protocol for a Pilot Clinical Trial. Nutrients 2023; 15:3068. [PMID: 37447394 PMCID: PMC10346691 DOI: 10.3390/nu15133068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
There is growing interest in the investigation of ketogenic diets as a potential therapy for bipolar disorder. The overlapping pharmacotherapies utilized for both bipolar disorder and seizures suggest that a mechanistic overlap may exist between these conditions, with fasting and the ketogenic diet representing the most time-proven therapies for seizure control. Recently, preliminary evidence has begun to emerge supporting a potential role for ketogenic diets in treating bipolar disorder. Notably, some patients may struggle to initiate a strict diet in the midst of a mood episode or significant life stressors. The key question addressed by this pilot clinical trial protocol is if benefits can be achieved with a less restrictive diet, as this would allow such an intervention to be accessible for more patients. Recent development of so-called ketone esters, that once ingested is converted to natural ketone bodies, combined with low glycemic index dietary changes has the potential to mimic two foundational components of therapeutic ketosis: high levels of ketones and minimal spiking of glucose/insulin. This pilot clinical trial protocol thus aims to investigate the effect of a 'ketogenic-mimicking diet' (combining supplementation of ketone esters with a low glycemic index dietary intervention) on neural network stability, mood, and biomarker outcomes in the setting of bipolar disorder. Positive findings obtained via this pilot clinical trial protocol may support future target engagement studies of ketogenic-mimicking diets or related ketogenic interventions. A lack of positive findings, in contrast, may justify a focus on more strict dietary interventions for future research.
Collapse
Affiliation(s)
| | | | - Alexis M. Griggs
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Stiven Roytman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | - Nicolaas I. Bohnen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Morris K. Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, MI 48109, USA
- Parkinson’s Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Melvin G. McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA
- Heinz C. Prechter Bipolar Research Program, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Leitner DF, Siu Y, Korman A, Lin Z, Kanshin E, Friedman D, Devore S, Ueberheide B, Tsirigos A, Jones DR, Wisniewski T, Devinsky O. Metabolomic, proteomic, and transcriptomic changes in adults with epilepsy on modified Atkins diet. Epilepsia 2023; 64:1046-1060. [PMID: 36775798 PMCID: PMC10372873 DOI: 10.1111/epi.17540] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/14/2023]
Abstract
OBJECTIVE High-fat and low-carbohydrate diets can reduce seizure frequency in some treatment-resistant epilepsy patients, including the more flexible modified Atkins diet (MAD), which is more palatable, mimicking fasting and inducing high ketone body levels. Low-carbohydrate diets may shift brain energy production, particularly impacting neuron- and astrocyte-linked metabolism. METHODS We evaluated the effect of short-term MAD on molecular mechanisms in adult epilepsy patients from surgical brain tissue and plasma compared to control participants consuming a nonmodified higher carbohydrate diet (n = 6 MAD, mean age = 43.7 years, range = 21-53, diet for average 10 days; n = 10 control, mean age = 41.9 years, range = 28-64). RESULTS By metabolomics, there were 13 increased metabolites in plasma (n = 15 participants with available specimens), which included 4.10-fold increased ketone body 3-hydroxybutyric acid, decreased palmitic acid in cortex (n = 16), and 11 decreased metabolites in hippocampus (n = 6), which had top associations with mitochondrial functions. Cortex and plasma 3-hydroxybutyric acid levels had a positive correlation (p = .0088, R2 = .48). Brain proteomics and RNAseq identified few differences, including 2.75-fold increased hippocampal MT-ND3 and trends (p < .01, false discovery rate > 5%) in hippocampal nicotinamide adenine dinucleotide (NADH)-related signaling pathways (activated oxidative phosphorylation and inhibited sirtuin signaling). SIGNIFICANCE Short-term MAD was associated with metabolic differences in plasma and resected epilepsy brain tissue when compared to control participants, in combination with trending expression changes observed in hippocampal NADH-related signaling pathways. Future studies should evaluate how brain molecular mechanisms are altered with long-term MAD in a larger cohort of epilepsy patients, with correlations to seizure frequency, epilepsy syndrome, and other clinical variables. [Clinicaltrials.gov NCT02565966.].
Collapse
Affiliation(s)
- Dominique F. Leitner
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, New York, United States of America
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Yik Siu
- Metabolomics Core Resource Laboratory, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Aryeh Korman
- Metabolomics Core Resource Laboratory, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Ziyan Lin
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Daniel Friedman
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Sasha Devore
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Beatrix Ueberheide
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Drew R. Jones
- Metabolomics Core Resource Laboratory, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Neurology, New York University Grossman School of Medicine, New York, New York, United States of America
| |
Collapse
|
8
|
Zhang Z, Bi X, Lian X, Niu Z. BDH1 promotes lung cancer cell proliferation and metastases by PARP1-mediated autophagy. J Cell Mol Med 2023; 27:939-949. [PMID: 36919822 PMCID: PMC10064033 DOI: 10.1111/jcmm.17700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 03/16/2023] Open
Abstract
Lymph node metastases and distant metastases were the important limiting factor for therapy of unresectable locally advanced (IIIB stage) and oligotransduction (IVa stage) lung cancer. This study was undertaken to identify a novel predictive biomarker for predicting lymph node metastases of lung cancer. A total of 364 patients with lung cancer which comprised of 198 patients with transcriptome sequencing data, 110 cases with immunohistochemistry data and 66 patients with serum samples were included to identify and validate the candidate gene. Autophagy was measured by western blots, immunofluorescence and electron microscope. We found that 3-hydroxybutyrate dehydrogenase 1 (BDH1) was associated with proliferation and metastases of lung cancer. BDH1 expression in both tissue and serum samples was associated with lung cancer metastases. Mechanical studies revealed that the AMPK-mTOR signalling pathway mediated by PARP1 played an important role in BDH1-induced autophagy. Activation of mTOR pathway markedly increased the effect of BDH1 in cell proliferation and metastases. These results were verified by the knockdown of PARP1. Furthermore, in vivo administration of BDH1 effectively promoted tumour growth in H460 xenografts mice. Our finding not only suggested that BDH1 might be useful as a novel biomarker and therapeutic target for lung cancer metastases, but also imply that PARP1-mediated AMPK-mTOR signalling pathway might play a critical role in BDH1-induced autophagy and lung cancer proliferation and metastases.
Collapse
Affiliation(s)
- Zhimin Zhang
- Thoracic Surgery, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Xin Bi
- Thoracic Surgery, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Xiaojuan Lian
- Department of Tumor Blood, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Zhongxi Niu
- Thoracic Surgery, The Third Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
9
|
Grande de França NA, Rolland Y, Guyonnet S, de Souto Barreto P. The role of dietary strategies in the modulation of hallmarks of aging. Ageing Res Rev 2023; 87:101908. [PMID: 36905962 DOI: 10.1016/j.arr.2023.101908] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
The hallmarks of aging constitute an interconnected network of basic mechanisms that modulate aging and can be modulated by lifestyle factors, including dietary strategies. This narrative review aimed to summarize the evidence on promoting dietary restriction or adherence to specific dietary patterns on hallmarks of aging. Studies with preclinical models or humans were considered. Dietary restriction (DR), usually operationalized as a reduction in caloric intake, is the main strategy applied to study the axis diet-hallmarks of aging. DR has been shown to modulate mainly genomic instability, loss of proteostasis, deregulating nutrient sensing, cellular senescence, and altered intercellular communication. Much less evidence exists on the role of dietary patterns, with most of the studies evaluating the Mediterranean Diet and other similar plant-based diets, and the ketogenic diet. Potential benefits are described in genomic instability, epigenetic alterations, loss of proteostasis, mitochondrial dysfunction, and altered intercellular communication. Given the predominant place of food in human life, it is imperative to determine the impact of nutritional strategies on the modulation of lifespan and healthspan, considering applicability, long-term adherence, and side effects.
Collapse
Affiliation(s)
- Natasha A Grande de França
- Gérontopôle of Toulouse, Institute on Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France.
| | - Yves Rolland
- Gérontopôle of Toulouse, Institute on Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; Maintain Aging Researchteam, CERPOP, Université de Toulouse, Inserm, Université Paul Sabatier, Toulouse, France
| | - Sophie Guyonnet
- Gérontopôle of Toulouse, Institute on Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; Maintain Aging Researchteam, CERPOP, Université de Toulouse, Inserm, Université Paul Sabatier, Toulouse, France
| | - Philipe de Souto Barreto
- Gérontopôle of Toulouse, Institute on Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; Maintain Aging Researchteam, CERPOP, Université de Toulouse, Inserm, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
10
|
Xu Y, Zheng F, Zhong Q, Zhu Y. Ketogenic Diet as a Promising Non-Drug Intervention for Alzheimer’s Disease: Mechanisms and Clinical Implications. J Alzheimers Dis 2023; 92:1173-1198. [PMID: 37038820 DOI: 10.3233/jad-230002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that is mainly characterized by cognitive deficits. Although many studies have been devoted to developing disease-modifying therapies, there has been no effective therapy until now. However, dietary interventions may be a potential strategy to treat AD. The ketogenic diet (KD) is a high-fat and low-carbohydrate diet with adequate protein. KD increases the levels of ketone bodies, providing an alternative energy source when there is not sufficient energy supply because of impaired glucose metabolism. Accumulating preclinical and clinical studies have shown that a KD is beneficial to AD. The potential underlying mechanisms include improved mitochondrial function, optimization of gut microbiota composition, and reduced neuroinflammation and oxidative stress. The review provides an update on clinical and preclinical research on the effects of KD or medium-chain triglyceride supplementation on symptoms and pathophysiology in AD. We also detail the potential mechanisms of KD, involving amyloid and tau proteins, neuroinflammation, gut microbiota, oxidative stress, and brain metabolism. We aimed to determine the function of the KD in AD and outline important aspects of the mechanism, providing a reference for the implementation of the KD as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Yunlong Xu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
- Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Fuxiang Zheng
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Qi Zhong
- Department of Neurology, Shenzhen Luohu People’s Hospital; The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Effect of the Ketone Body, D-β-Hydroxybutyrate, on Sirtuin2-Mediated Regulation of Mitochondrial Quality Control and the Autophagy-Lysosomal Pathway. Cells 2023; 12:cells12030486. [PMID: 36766827 PMCID: PMC9914182 DOI: 10.3390/cells12030486] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Mitochondrial activity and quality control are essential for neuronal homeostasis as neurons rely on glucose oxidative metabolism. The ketone body, D-β-hydroxybutyrate (D-BHB), is metabolized to acetyl-CoA in brain mitochondria and used as an energy fuel alternative to glucose. We have previously reported that D-BHB sustains ATP production and stimulates the autophagic flux under glucose deprivation in neurons; however, the effects of D-BHB on mitochondrial turnover under physiological conditions are still unknown. Sirtuins (SIRTs) are NAD+-activated protein deacetylases involved in the regulation of mitochondrial biogenesis and mitophagy through the activation of transcription factors FOXO1, FOXO3a, TFEB and PGC1α coactivator. Here, we aimed to investigate the effect of D-BHB on mitochondrial turnover in cultured neurons and the mechanisms involved. Results show that D-BHB increased mitochondrial membrane potential and regulated the NAD+/NADH ratio. D-BHB enhanced FOXO1, FOXO3a and PGC1α nuclear levels in an SIRT2-dependent manner and stimulated autophagy, mitophagy and mitochondrial biogenesis. These effects increased neuronal resistance to energy stress. D-BHB also stimulated the autophagic-lysosomal pathway through AMPK activation and TFEB-mediated lysosomal biogenesis. Upregulation of SIRT2, FOXOs, PGC1α and TFEB was confirmed in the brain of ketogenic diet (KD)-treated mice. Altogether, the results identify SIRT2, for the first time, as a target of D-BHB in neurons, which is involved in the regulation of autophagy/mitophagy and mitochondrial quality control.
Collapse
|
12
|
Wang Y, Zhang J, Zhang Y, Yao J. Bibliometric analysis of global research profile on ketogenic diet therapies in neurological diseases: Beneficial diet therapies deserve more attention. Front Endocrinol (Lausanne) 2023; 13:1066785. [PMID: 36686482 PMCID: PMC9846225 DOI: 10.3389/fendo.2022.1066785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Background The protective effects of Ketogenic Diet Therapies (KDTs) on neurological diseases have been extensively studied over the past two decades. The purpose of this study was to quantitatively and qualitatively analyze the publication of KDTs in the neurological field from 2000 to 2021. Methods A literature search was performed on June 7th, 2022, using the search terms: (("ketone" OR "ketogenic" OR "*hydroxybuty*") AND ("neuro*")) in the WoSCC database. Collected data were further analyzed using VOSviewer, CiteSpace and other online bibliometric websites. The annual publication volume and citation trends were summarized. The collaborations among highly cited countries, institutions, authors and journals were visualized. The co-citation analysis of highly cited references and journals were also visualized. Moreover, the research focuses and fronts were revealed by co-occurrence analysis and burst keywords detection. Results A total of 2808 publications with 88,119 citations were identified. From 2000-2021, the number of publications and citations presented rising trends. The United States was the country with an overwhelming number of publications and cited times. Johns Hopkins University was the most contributory institution. Kossoff Eric H was the author with the largest number of publications. And Epilepsia was both the largest publisher and the most frequently cited journal. The keywords of intense interest involved "Modified Atkins Diet", "Temporal Lobe Epilepsy", "Alzheimer's Disease", "Parkinson's Disease", "Cerebral Blood Flow", "Neuroinflammation", "Oxidative Stress", "Metabolism" and "Mitochondria". Conclusion We presented the global trend of KDTs in neurological diseases and provided important information for relevant researchers in a bibliometric way. This bibliometric study revealed that treating epilepsy, neuroprotection and functional effects of KDTs on mitochondria and oxidative stress have been the spotlight from 2000 to 2021. These have emerged as the basis for transformation from basic research to clinical application of KDTs.
Collapse
Affiliation(s)
| | | | | | - Junyan Yao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Tozzi R, Campolo F, Baldini E, Venneri MA, Lubrano C, Ulisse S, Gnessi L, Mariani S. Ketogenic Diet Increases Serum and White Adipose Tissue SIRT1 Expression in Mice. Int J Mol Sci 2022; 23:ijms232415860. [PMID: 36555502 PMCID: PMC9785229 DOI: 10.3390/ijms232415860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Overnutrition and its sequelae have become a global concern due to the increasing incidence of obesity and insulin resistance. A ketogenic diet (KD) is widely used as a dietary treatment for metabolic disorders. Sirtuin1 (SIRT1), a metabolic sensor which regulates fat homeostasis, is modulated by dietary interventions. However, the influence of nutritional ketosis on SIRT1 is still debated. We examined the effect of KD on adipose tissue, liver, and serum levels of SIRT1 in mice. Adult C57BL/6J male mice were randomly assigned to two isocaloric dietary groups and fed with either high-fat KD or normal chow (NC) for 4 weeks. Serum SIRT1, beta-hydroxybutyrate (βHB), glucose, and triglyceride levels, as well as SIRT1 expression in visceral (VAT), subcutaneous (SAT), and brown (BAT) adipose tissues, and in the liver, were measured. KD-fed mice showed an increase in serum βHB in parallel with serum SIRT1 (r = 0.732, p = 0.0156), and increased SIRT1 protein expression in SAT and VAT. SIRT1 levels remained unchanged in BAT and in the liver, which developed steatosis. Normal glycemia and triglycerides were observed. Under a KD, serum and white fat phenotypes show higher SIRT1, suggesting that one of the molecular mechanisms underlying a KD's potential benefits on metabolic health involves a synergistic interaction with SIRT1.
Collapse
Affiliation(s)
- Rossella Tozzi
- Department of Molecular Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Enke Baldini
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Carla Lubrano
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Salvatore Ulisse
- Department of Surgical Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefania Mariani
- Department of Experimental Medicine, Section of Medical Physiopathology, Food Science and Endocrinology, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
- Correspondence: ; Tel.: +39-6-49970509; Fax: +39-6-4461450
| |
Collapse
|
14
|
Aminzadeh-Gohari S, Kofler B, Herzog C. Dietary restriction in senolysis and prevention and treatment of disease. Crit Rev Food Sci Nutr 2022; 64:5242-5268. [PMID: 36484738 PMCID: PMC7616065 DOI: 10.1080/10408398.2022.2153355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aging represents a key risk factor for a plethora of diseases. Targeting detrimental processes which occur during aging, especially before onset of age-related disease, could provide drastic improvements in healthspan. There is increasing evidence that dietary restriction (DR), including caloric restriction, fasting, or fasting-mimicking diets, extend both lifespan and healthspan. This has sparked interest in the use of dietary regimens as a non-pharmacological means to slow aging and prevent disease. Here, we review the current evidence on the molecular mechanisms underlying DR-induced health improvements, including removal of senescent cells, metabolic reprogramming, and epigenetic rejuvenation.
Collapse
Affiliation(s)
- Sepideh Aminzadeh-Gohari
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Lkhagva B, Lee TW, Lin YK, Chen YC, Chung CC, Higa S, Chen YJ. Disturbed Cardiac Metabolism Triggers Atrial Arrhythmogenesis in Diabetes Mellitus: Energy Substrate Alternate as a Potential Therapeutic Intervention. Cells 2022; 11:cells11182915. [PMID: 36139490 PMCID: PMC9497243 DOI: 10.3390/cells11182915] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of sustained arrhythmia in diabetes mellitus (DM). Its morbidity and mortality rates are high, and its prevalence will increase as the population ages. Despite expanding knowledge on the pathophysiological mechanisms of AF, current pharmacological interventions remain unsatisfactory; therefore, novel findings on the underlying mechanism are required. A growing body of evidence suggests that an altered energy metabolism is closely related to atrial arrhythmogenesis, and this finding engenders novel insights into the pathogenesis of the pathophysiology of AF. In this review, we provide comprehensive information on the mechanistic insights into the cardiac energy metabolic changes, altered substrate oxidation rates, and mitochondrial dysfunctions involved in atrial arrhythmogenesis, and suggest a promising advanced new therapeutic approach to treat patients with AF.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| | - Cheng-Chih Chung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa 901-2131, Japan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cardiovascular Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Correspondence:
| |
Collapse
|
16
|
Ketone Bodies and SIRT1, Synergic Epigenetic Regulators for Metabolic Health: A Narrative Review. Nutrients 2022; 14:nu14153145. [PMID: 35956321 PMCID: PMC9370141 DOI: 10.3390/nu14153145] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Ketone bodies (KBs) and Sirtuin-1 (SIRT1) have received increasing attention over the past two decades given their pivotal function in a variety of biological contexts, including transcriptional regulation, cell cycle progression, inflammation, metabolism, neurological and cardiovascular physiology, and cancer. As a consequence, the modulation of KBs and SIRT1 is considered a promising therapeutic option for many diseases. The direct regulation of gene expression can occur in vivo through histone modifications mediated by both SIRT1 and KBs during fasting or low-carbohydrate diets, and dietary metabolites may contribute to epigenetic regulation, leading to greater genomic plasticity. In this review, we provide an updated overview of the epigenetic interactions between KBs and SIRT1, with a particular glance at their central, synergistic roles for metabolic health.
Collapse
|
17
|
Taylor MK, Sullivan DK, Keller JE, Burns JM, Swerdlow RH. Potential for Ketotherapies as Amyloid-Regulating Treatment in Individuals at Risk for Alzheimer’s Disease. Front Neurosci 2022; 16:899612. [PMID: 35784855 PMCID: PMC9243383 DOI: 10.3389/fnins.2022.899612] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/30/2022] [Indexed: 12/27/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative condition characterized by clinical decline in memory and other cognitive functions. A classic AD neuropathological hallmark includes the accumulation of amyloid-β (Aβ) plaques, which may precede onset of clinical symptoms by over a decade. Efforts to prevent or treat AD frequently emphasize decreasing Aβ through various mechanisms, but such approaches have yet to establish compelling interventions. It is still not understood exactly why Aβ accumulates in AD, but it is hypothesized that Aβ and other downstream pathological events are a result of impaired bioenergetics, which can also manifest prior to cognitive decline. Evidence suggests that individuals with AD and at high risk for AD have functional brain ketone metabolism and ketotherapies (KTs), dietary approaches that produce ketone bodies for energy metabolism, may affect AD pathology by targeting impaired brain bioenergetics. Cognitively normal individuals with elevated brain Aβ, deemed “preclinical AD,” and older adults with peripheral metabolic impairments are ideal candidates to test whether KTs modulate AD biology as they have impaired mitochondrial function, perturbed brain glucose metabolism, and elevated risk for rapid Aβ accumulation and symptomatic AD. Here, we discuss the link between brain bioenergetics and Aβ, as well as the potential for KTs to influence AD risk and progression.
Collapse
Affiliation(s)
- Matthew K. Taylor
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
- *Correspondence: Matthew K. Taylor,
| | - Debra K. Sullivan
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
| | - Jessica E. Keller
- Department of Dietetics and Nutrition, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
18
|
Increased Hippocampal Afterdischarge Threshold in Ketogenic Diet is Accompanied by Enhanced Kynurenine Pathway Activity. Neurochem Res 2022; 47:2109-2122. [PMID: 35522366 DOI: 10.1007/s11064-022-03605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/22/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
The efficacy of a ketogenic diet (KD) in controlling seizure has been shown in many experimental and clinical studies, however, its mechanism of action still needs further clarification. The major goal of the present study was to investigate the influence of the commercially available KD and caloric restriction (CR) on the hippocampal afterdischarge (AD) threshold in rats, and concomitant biochemical changes, specifically concerning the kynurenine pathway, in plasma and the hippocampus. As expected, the rats on the KD showed higher AD threshold accompanied by increased plasma β-hydroxybutyrate level compared to the control group and the CR rats. This group presented also lowered tryptophan and elevated kynurenic acid levels in plasma with similar changes in the hippocampus. Moreover, the KD rats showed decreased levels of branched chain amino acids (BCAA) and aromatic amino acids (AAA) in plasma and the hippocampus. No regular biochemical changes were observed in the CR group. Our results are analogous to those detected after single administrations of fatty acids and valproic acid in our previous studies, specifically to an increase in the kynurenine pathway activity and changes in peripheral and central BCAA and AAA levels. This suggests that the anticonvulsant effect of the KD may be at least partially associated with those observed biochemical alternations.
Collapse
|
19
|
Kim CK, Sachdev PS, Braidy N. Recent Neurotherapeutic Strategies to Promote Healthy Brain Aging: Are we there yet? Aging Dis 2022; 13:175-214. [PMID: 35111369 PMCID: PMC8782556 DOI: 10.14336/ad.2021.0705] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Owing to the global exponential increase in population ageing, there is an urgent unmet need to develop reliable strategies to slow down and delay the ageing process. Age-related neurodegenerative diseases are among the main causes of morbidity and mortality in our contemporary society and represent a major socio-economic burden. There are several controversial factors that are thought to play a causal role in brain ageing which are continuously being examined in experimental models. Among them are oxidative stress and brain inflammation which are empirical to brain ageing. Although some candidate drugs have been developed which reduce the ageing phenotype, their clinical translation is limited. There are several strategies currently in development to improve brain ageing. These include strategies such as caloric restriction, ketogenic diet, promotion of cellular nicotinamide adenine dinucleotide (NAD+) levels, removal of senescent cells, 'young blood' transfusions, enhancement of adult neurogenesis, stem cell therapy, vascular risk reduction, and non-pharmacological lifestyle strategies. Several studies have shown that these strategies can not only improve brain ageing by attenuating age-related neurodegenerative disease mechanisms, but also maintain cognitive function in a variety of pre-clinical experimental murine models. However, clinical evidence is limited and many of these strategies are awaiting findings from large-scale clinical trials which are nascent in the current literature. Further studies are needed to determine their long-term efficacy and lack of adverse effects in various tissues and organs to gain a greater understanding of their potential beneficial effects on brain ageing and health span in humans.
Collapse
Affiliation(s)
- Chul-Kyu Kim
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Perminder S Sachdev
- Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
20
|
Avgerinos KI, Mullins RJ, Egan JM, Kapogiannis D. Ketone Ester Effects on Biomarkers of Brain Metabolism and Cognitive Performance in Cognitively Intact Adults ≥ 55 Years Old. A Study Protocol for a Double-Blinded Randomized Controlled Clinical Trial. J Prev Alzheimers Dis 2022; 9:54-66. [PMID: 35098974 PMCID: PMC9359666 DOI: 10.14283/jpad.2022.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ketone bodies have been proposed as an "energy rescue" for the Alzheimer's disease (AD) brain, which underutilizes glucose. Prior research has shown that oral ketone monoester (KME) safely induces robust ketosis in humans and has demonstrated cognitive-enhancing and pathology-reducing properties in animal models of AD. However, human evidence that KME may enhance brain ketone metabolism, improve cognitive performance and engage AD pathogenic cascades is scarce. OBJECTIVES To investigate the effects of ketone monoester (KME) on brain metabolism, cognitive performance and AD pathogenic cascades in cognitively normal older adults with metabolic syndrome and therefore at higher risk for AD. DESIGN Double-blinded randomized placebo-controlled clinical trial. SETTING Clinical Unit of the National Institute on Aging, Baltimore, US. PARTICIPANTS Fifty cognitively intact adults ≥ 55 years old, with metabolic syndrome. INTERVENTION Drinks containing 25 g of KME or isocaloric placebo consumed three times daily for 28 days. OUTCOMES Primary: concentration of beta-hydroxybutyrate (BHB) in precuneus measured with Magnetic Resonance Spectroscopy (MRS). Exploratory: plasma and urine BHB, multiple brain and muscle metabolites detected with MRS, cognition assessed with the PACC and NIH toolbox, biomarkers of AD and metabolic mediators in plasma extracellular vesicles, and stool microbiome. DISCUSSION This is the first study to investigate the AD-biomarker and cognitive effects of KME in humans. Ketone monoester is safe, tolerable, induces robust ketosis, and animal studies indicate that it can modify AD pathology. By conducting a study of KME in a population at risk for AD, we hope to bridge the existing gap between pre-clinical evidence and the potential for brain-metabolic, pro-cognitive, and anti-Alzheimer's effects in humans.
Collapse
Affiliation(s)
- K I Avgerinos
- Dimitrios Kapogiannis, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, 8C228, Baltimore, MD 21224, USA; Email address: ; Telephone: +1 410 350 3953
| | | | | | | |
Collapse
|
21
|
Ketogenic diets and the nervous system: a scoping review of neurological outcomes from nutritional ketosis in animal studies. Nutr Res Rev 2021; 35:268-281. [PMID: 34180385 DOI: 10.1017/s0954422421000214] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Ketogenic diets have reported efficacy for neurological dysfunctions; however, there are limited published human clinical trials elucidating the mechanisms by which nutritional ketosis produces therapeutic effects. The purpose of this present study was to investigate animal models that report variations in nervous system function by changing from a standard animal diet to a ketogenic diet, synthesise these into broad themes, and compare these with mechanisms reported as targets in pain neuroscience to inform human chronic pain trials. METHODS An electronic search of seven databases was conducted in July 2020. Two independent reviewers screened studies for eligibility, and descriptive outcomes relating to nervous system function were extracted for a thematic analysis, then synthesised into broad themes. RESULTS In total, 170 studies from eighteen different disease models were identified and grouped into fourteen broad themes: alterations in cellular energetics and metabolism, biochemical, cortical excitability, epigenetic regulation, mitochondrial function, neuroinflammation, neuroplasticity, neuroprotection, neurotransmitter function, nociception, redox balance, signalling pathways, synaptic transmission and vascular supply. DISCUSSION The mechanisms presented centred around the reduction of inflammation and oxidative stress as well as a reduction in nervous system excitability. Given the multiple potential mechanisms presented, it is likely that many of these are involved synergistically and undergo adaptive processes within the human body, and controlled animal models that limit the investigation to a particular pathway in isolation may reach differing conclusions. Attention is required when translating this information to human chronic pain populations owing to the limitations outlined from the animal research.
Collapse
|
22
|
Eat, Train, Sleep-Retreat? Hormonal Interactions of Intermittent Fasting, Exercise and Circadian Rhythm. Biomolecules 2021; 11:biom11040516. [PMID: 33808424 PMCID: PMC8065500 DOI: 10.3390/biom11040516] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 01/08/2023] Open
Abstract
The circadian rhythmicity of endogenous metabolic and hormonal processes is controlled by a complex system of central and peripheral pacemakers, influenced by exogenous factors like light/dark-cycles, nutrition and exercise timing. There is evidence that alterations in this system may be involved in the pathogenesis of metabolic diseases. It has been shown that disruptions to normal diurnal rhythms lead to drastic changes in circadian processes, as often seen in modern society due to excessive exposure to unnatural light sources. Out of that, research has focused on time-restricted feeding and exercise, as both seem to be able to reset disruptions in circadian pacemakers. Based on these results and personal physical goals, optimal time periods for food intake and exercise have been identified. This review shows that appropriate nutrition and exercise timing are powerful tools to support, rather than not disturb, the circadian rhythm and potentially contribute to the prevention of metabolic diseases. Nevertheless, both lifestyle interventions are unable to address the real issue: the misalignment of our biological with our social time.
Collapse
|
23
|
Liśkiewicz D, Liśkiewicz A, Grabowski M, Nowacka-Chmielewska MM, Jabłońska K, Wojakowska A, Marczak Ł, Barski JJ, Małecki A. Upregulation of hepatic autophagy under nutritional ketosis. J Nutr Biochem 2021; 93:108620. [PMID: 33705944 DOI: 10.1016/j.jnutbio.2021.108620] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/15/2020] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
Many of the metabolic effects evoked by the ketogenic diet mimic the actions of fasting and the benefits of the ketogenic diet are often attributed to these similarities. Since fasting is a potent autophagy inductor in vivo and in vitro it has been hypothesized that the ketogenic diet may upregulate autophagy. The aim of the present study was to provide a comprehensive evaluation of the influence of the ketogenic diet on the hepatic autophagy. C57BL/6N male mice were fed with two different ketogenic chows composed of fat of either animal or plant origin for 4 weeks. To gain some insight into the time frame for the induction of autophagy on the ketogenic diet, we performed a short-term experiment in which animals were fed with ketogenic diets for only 24 or 48 h. The results showed that autophagy is upregulated in the livers of animals fed with the ketogenic diet. Moreover, the size of the observed effect was likely dependent on the diet composition. Subsequently, the markers of regulatory pathways that may link ketogenic diet action to autophagy were measured, i.e., the activity of mTORC1, activation of AMPK, and the levels of SIRT1, p53, and FOXO3. Overall, observed treatment-specific effects including the upregulation of SIRT1 and downregulation of FOXO3 and p53. Finally, a GC/MS analysis of the fatty acid composition of animals' livers and the chows was performed in order to obtain an idea about the presence of specific compounds that may shape the effects of ketogenic diets on autophagy.
Collapse
Affiliation(s)
- Daniela Liśkiewicz
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
| | - Arkadiusz Liśkiewicz
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Mateusz Grabowski
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marta Maria Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Konstancja Jabłońska
- Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Anna Wojakowska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Łukasz Marczak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Jarosław J Barski
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland; Department for Experimental Medicine, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Andrzej Małecki
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| |
Collapse
|
24
|
Masino SA, Ruskin DN, Freedgood NR, Lindefeldt M, Dahlin M. Differential ketogenic diet-induced shift in CSF lipid/carbohydrate metabolome of pediatric epilepsy patients with optimal vs. no anticonvulsant response: a pilot study. Nutr Metab (Lond) 2021; 18:23. [PMID: 33648550 PMCID: PMC7923458 DOI: 10.1186/s12986-020-00524-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/21/2020] [Indexed: 02/02/2023] Open
Abstract
Background The low carbohydrate, high fat ketogenic diet can be an effective anticonvulsant treatment in some pediatric patients with pharmacoresistant epilepsy. Its mechanism(s) of action, however, remain uncertain. Direct sampling of cerebrospinal fluid before and during metabolic therapy may reveal key changes associated with differential clinical outcomes. We characterized the relationship between seizure responsiveness and changes in lipid and carbohydrate metabolites. Methods We performed metabolomic analysis of cerebrospinal fluid samples taken before and during ketogenic diet treatment in patients with optimal response (100% seizure remission) and patients with no response (no seizure improvement) to search for differential diet effects in hallmark metabolic compounds in these two groups. Optimal responders and non-responders were similar in age range and included males and females. Seizure types and the etiologies or syndromes of epilepsy varied but did not appear to differ systematically between responders and non-responders. Results Analysis showed a strong effect of ketogenic diet treatment on the cerebrospinal fluid metabolome. Longitudinal and between-subjects analyses revealed that many lipids and carbohydrates were changed significantly by ketogenic diet, with changes typically being of larger magnitude in responders. Notably, responders had more robust changes in glucose and the ketone bodies β-hydroxybutyrate and acetoacetate than non-responders; conversely, non-responders had significant increases in fructose and sorbose, which did not occur in responders. Conclusions The data suggest that a differential and stronger metabolic response to the ketogenic diet may predict a better anticonvulsant response, and such variability is likely due to inherent biological factors of individual patients. Strategies to boost the metabolic response may be beneficial.
Collapse
Affiliation(s)
- Susan A Masino
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, CT, 06106, USA
| | - David N Ruskin
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, CT, 06106, USA.
| | - Natalie R Freedgood
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, CT, 06106, USA
| | - Marie Lindefeldt
- Neuropediatric Department, Astrid Lindgren Children's Hospital, Karolinska Hospital, Stockholm, Sweden
| | - Maria Dahlin
- Neuropediatric Department, Astrid Lindgren Children's Hospital, Karolinska Hospital, Stockholm, Sweden
| |
Collapse
|
25
|
Ruskin DN, Sturdevant IC, Wyss LS, Masino SA. Ketogenic diet effects on inflammatory allodynia and ongoing pain in rodents. Sci Rep 2021; 11:725. [PMID: 33436956 PMCID: PMC7804255 DOI: 10.1038/s41598-020-80727-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022] Open
Abstract
Ketogenic diets are very low carbohydrate, high fat, moderate protein diets used to treat medication-resistant epilepsy. Growing evidence suggests that one of the ketogenic diet’s main mechanisms of action is reducing inflammation. Here, we examined the diet’s effects on experimental inflammatory pain in rodent models. Young adult rats and mice were placed on the ketogenic diet or maintained on control diet. After 3–4 weeks on their respective diets, complete Freund’s adjuvant (CFA) was injected in one hindpaw to induce inflammation; the contralateral paw was used as the control. Tactile sensitivity (von Frey) and indicators of spontaneous pain were quantified before and after CFA injection. Ketogenic diet treatment significantly reduced tactile allodynia in both rats and mice, though with a species-specific time course. There was a strong trend to reduced spontaneous pain in rats but not mice. These data suggest that ketogenic diets or other ketogenic treatments might be useful treatments for conditions involving inflammatory pain.
Collapse
Affiliation(s)
- David N Ruskin
- Neuroscience Program and Department of Psychology, Trinity College, 300 Summit St., Hartford, CT, 06106, USA.
| | - Isabella C Sturdevant
- Neuroscience Program and Department of Psychology, Trinity College, 300 Summit St., Hartford, CT, 06106, USA
| | - Livia S Wyss
- Neuroscience Program and Department of Psychology, Trinity College, 300 Summit St., Hartford, CT, 06106, USA
| | - Susan A Masino
- Neuroscience Program and Department of Psychology, Trinity College, 300 Summit St., Hartford, CT, 06106, USA
| |
Collapse
|
26
|
Mahajan VR, Elvig SK, Vendruscolo LF, Koob GF, Darcey VL, King MT, Kranzler HR, Volkow ND, Wiers CE. Nutritional Ketosis as a Potential Treatment for Alcohol Use Disorder. Front Psychiatry 2021; 12:781668. [PMID: 34916977 PMCID: PMC8670944 DOI: 10.3389/fpsyt.2021.781668] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/08/2021] [Indexed: 12/28/2022] Open
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing brain disorder, characterized by compulsive alcohol seeking and disrupted brain function. In individuals with AUD, abstinence from alcohol often precipitates withdrawal symptoms than can be life threatening. Here, we review evidence for nutritional ketosis as a potential means to reduce withdrawal and alcohol craving. We also review the underlying mechanisms of action of ketosis. Several findings suggest that during alcohol intoxication there is a shift from glucose to acetate metabolism that is enhanced in individuals with AUD. During withdrawal, there is a decline in acetate levels that can result in an energy deficit and could contribute to neurotoxicity. A ketogenic diet or ingestion of a ketone ester elevates ketone bodies (acetoacetate, β-hydroxybutyrate and acetone) in plasma and brain, resulting in nutritional ketosis. These effects have been shown to reduce alcohol withdrawal symptoms, alcohol craving, and alcohol consumption in both preclinical and clinical studies. Thus, nutritional ketosis may represent a unique treatment option for AUD: namely, a nutritional intervention that could be used alone or to augment the effects of medications.
Collapse
Affiliation(s)
- Vikrant R Mahajan
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Sophie K Elvig
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Leandro F Vendruscolo
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - George F Koob
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Valerie L Darcey
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - M Todd King
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, United States
| | - Henry R Kranzler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, United States
| | - Corinde E Wiers
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
27
|
From Obesity to Hippocampal Neurodegeneration: Pathogenesis and Non-Pharmacological Interventions. Int J Mol Sci 2020; 22:ijms22010201. [PMID: 33379163 PMCID: PMC7796248 DOI: 10.3390/ijms22010201] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/16/2022] Open
Abstract
High-caloric diet and physical inactivity predispose individuals to obesity and diabetes, which are risk factors of hippocampal neurodegeneration and cognitive deficits. Along with the adipose-hippocampus crosstalk, chronically inflamed adipose tissue secretes inflammatory cytokine could trigger neuroinflammatory responses in the hippocampus, and in turn, impairs hippocampal neuroplasticity under obese and diabetic conditions. Hence, caloric restriction and physical exercise are critical non-pharmacological interventions to halt the pathogenesis from obesity to hippocampal neurodegeneration. In response to physical exercise, peripheral organs, including the adipose tissue, skeletal muscles, and liver, can secret numerous exerkines, which bring beneficial effects to metabolic and brain health. In this review, we summarized how chronic inflammation in adipose tissue could trigger neuroinflammation and hippocampal impairment, which potentially contribute to cognitive deficits in obese and diabetic conditions. We also discussed the potential mechanisms underlying the neurotrophic and neuroprotective effects of caloric restriction and physical exercise by counteracting neuroinflammation, plasticity deficits, and cognitive impairments. This review provides timely insights into how chronic metabolic disorders, like obesity, could impair brain health and cognitive functions in later life.
Collapse
|
28
|
Covarrubias AJ, Perrone R, Grozio A, Verdin E. NAD + metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol 2020; 22:119-141. [PMID: 33353981 DOI: 10.1038/s41580-020-00313-x] [Citation(s) in RCA: 673] [Impact Index Per Article: 168.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a coenzyme for redox reactions, making it central to energy metabolism. NAD+ is also an essential cofactor for non-redox NAD+-dependent enzymes, including sirtuins, CD38 and poly(ADP-ribose) polymerases. NAD+ can directly and indirectly influence many key cellular functions, including metabolic pathways, DNA repair, chromatin remodelling, cellular senescence and immune cell function. These cellular processes and functions are critical for maintaining tissue and metabolic homeostasis and for healthy ageing. Remarkably, ageing is accompanied by a gradual decline in tissue and cellular NAD+ levels in multiple model organisms, including rodents and humans. This decline in NAD+ levels is linked causally to numerous ageing-associated diseases, including cognitive decline, cancer, metabolic disease, sarcopenia and frailty. Many of these ageing-associated diseases can be slowed down and even reversed by restoring NAD+ levels. Therefore, targeting NAD+ metabolism has emerged as a potential therapeutic approach to ameliorate ageing-related disease, and extend the human healthspan and lifespan. However, much remains to be learnt about how NAD+ influences human health and ageing biology. This includes a deeper understanding of the molecular mechanisms that regulate NAD+ levels, how to effectively restore NAD+ levels during ageing, whether doing so is safe and whether NAD+ repletion will have beneficial effects in ageing humans.
Collapse
Affiliation(s)
- Anthony J Covarrubias
- Buck Institute for Research on Aging, Novato, CA, USA.,UCSF Department of Medicine, San Francisco, CA, USA
| | | | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA. .,UCSF Department of Medicine, San Francisco, CA, USA.
| |
Collapse
|
29
|
Elamin M, Ruskin DN, Sacchetti P, Masino SA. A unifying mechanism of ketogenic diet action: The multiple roles of nicotinamide adenine dinucleotide. Epilepsy Res 2020; 167:106469. [PMID: 33038721 DOI: 10.1016/j.eplepsyres.2020.106469] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/22/2020] [Accepted: 09/09/2020] [Indexed: 01/23/2023]
Abstract
The ability of a ketogenic diet to treat seizures and render a neuronal network more resistant to strong electrical activity has been observed for a century in clinics and for decades in research laboratories. Alongside ongoing efforts to understand how this therapy works to stop seizures, metabolic health is increasingly appreciated as critical buffer to resisting and recovering from acute and chronic disease. Accordingly, links between metabolism and health, and the broader emerging impact of the ketogenic diet in improving diverse metabolic, immunological and neurological conditions, have served to intensify the search for its key and/or common mechanisms. Here we review diverse evidence for increased levels of NAD+, and thus an altered ratio of NAD+/NADH, during metabolic therapy with a ketogenic diet. We propose this as a potential unifying mechanism, and highlight some of the evidence linking altered NAD+/NADH with reduced seizures and with a range of short and long-term changes associated with the beneficial effects of a ketogenic diet. An increase in NAD+/NADH is consistent with multiple lines of evidence and hypotheses, and therefore we suggest that increased NAD+ may be a common mechanism underlying beneficial effects of ketogenic diet therapy.
Collapse
Affiliation(s)
- Marwa Elamin
- Neuroscience Department, UConn School of Medicine, Farmington CT, United States.
| | - David N Ruskin
- Neuroscience Program & Psychology Department, Trinity College, Hartford, CT, United States.
| | - Paola Sacchetti
- Neuroscience Program & Department of Biology, University of Hartford, West Hartford, CT, United States.
| | - Susan A Masino
- Neuroscience Program & Psychology Department, Trinity College, Hartford, CT, United States.
| |
Collapse
|
30
|
Morris G, Puri BK, Maes M, Olive L, Berk M, Carvalho AF. The role of microglia in neuroprogressive disorders: mechanisms and possible neurotherapeutic effects of induced ketosis. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109858. [PMID: 31923453 DOI: 10.1016/j.pnpbp.2020.109858] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 12/23/2022]
Abstract
A comprehensive review of molecular mechanisms involved in the promotion and maintenance of distinct microglia phenotypes is provided. The acquisition and perpetuation of predominantly pro-inflammatory microglial phenotypes have been implicated in the pathophysiology of several neuroprogressive diseases and is associated with reduced ATP production via oxidative phosphorylation, increased ATP generation by glycolysis, elevated oxidative and nitrosative stress and other metabolic, inflammatory and hormonal insults. Microglia can also adopt a predominantly anti-inflammatory phenotypes with neuroprotective properties. Strategies that promote and maintain a predominantly anti-inflammatory phenotype may hold promise as novel therapeutic opportunities for neuroprogressive illness. Induced ketosis may promote a transition towards predominantly anti-inflammatory microglial states/phenotypes by several mechanisms, including inhibition of glycolysis and increased NAD+ production; engagement of microglial GPR109A receptors; histone deacetylase inhibition; and elevated n-3 polyunsaturated fatty acids levels. Since microglia activation can now be assessed in vivo, these data provide a clear rationale for the design of transdiagnostic randomized controlled trials of the ketogenic diet and other ketosis-inducing strategies for neuroprogressive diseases, which may also provide mechanistic insights through the assessment of "target engagement".
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | | | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Lisa Olive
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Andre F Carvalho
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.
| |
Collapse
|
31
|
Morris G, Puri BK, Carvalho A, Maes M, Berk M, Ruusunen A, Olive L. Induced Ketosis as a Treatment for Neuroprogressive Disorders: Food for Thought? Int J Neuropsychopharmacol 2020; 23:366-384. [PMID: 32034911 PMCID: PMC7311648 DOI: 10.1093/ijnp/pyaa008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/05/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Induced ketosis (or ketone body ingestion) can ameliorate several changes associated with neuroprogressive disorders, including schizophrenia, bipolar disorder, and major depressive disorder. Thus, the effects of glucose hypometabolism can be bypassed through the entry of beta-hydroxybutyrate, providing an alternative source of energy to glucose. The weight of evidence suggests that induced ketosis reduces levels of oxidative stress, mitochondrial dysfunction, and inflammation-core features of the above disorders. There are also data to suggest that induced ketosis may be able to target other molecules and signaling pathways whose levels and/or activity are also known to be abnormal in at least some patients suffering from these illnesses such as peroxisome proliferator-activated receptors, increased activity of the Kelch-like ECH-associated protein/nuclear factor erythroid 2-related factor 2, Sirtuin-1 nuclear factor-κB p65, and nicotinamide adenine dinucleotide (NAD). This review explains the mechanisms by which induced ketosis might reduce mitochondrial dysfunction, inflammation, and oxidative stress in neuropsychiatric disorders and ameliorate abnormal levels of molecules and signaling pathways that also appear to contribute to the pathophysiology of these illnesses. This review also examines safety data relating to induced ketosis over the long term and discusses the design of future studies.
Collapse
Affiliation(s)
- Gerwyn Morris
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia
| | - Basant K Puri
- C.A.R., Cambridge, United Kingdom,Hammersmith Hospital, London, United Kingdom
| | - Andre Carvalho
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Michael Maes
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria,Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia,Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry, and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia,Correspondence: Michael Berk, PO Box 281 Geelong, Victoria 3220 Australia ()
| | - Anu Ruusunen
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia
| | - Lisa Olive
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Australia
| |
Collapse
|
32
|
Morris G, Maes M, Berk M, Carvalho AF, Puri BK. Nutritional ketosis as an intervention to relieve astrogliosis: Possible therapeutic applications in the treatment of neurodegenerative and neuroprogressive disorders. Eur Psychiatry 2020; 63:e8. [PMID: 32093791 PMCID: PMC8057392 DOI: 10.1192/j.eurpsy.2019.13] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Nutritional ketosis, induced via either the classical ketogenic diet or the use of emulsified medium-chain triglycerides, is an established treatment for pharmaceutical resistant epilepsy in children and more recently in adults. In addition, the use of oral ketogenic compounds, fractionated coconut oil, very low carbohydrate intake, or ketone monoester supplementation has been reported to be potentially helpful in mild cognitive impairment, Parkinson’s disease, schizophrenia, bipolar disorder, and autistic spectrum disorder. In these and other neurodegenerative and neuroprogressive disorders, there are detrimental effects of oxidative stress, mitochondrial dysfunction, and neuroinflammation on neuronal function. However, they also adversely impact on neurone–glia interactions, disrupting the role of microglia and astrocytes in central nervous system (CNS) homeostasis. Astrocytes are the main site of CNS fatty acid oxidation; the resulting ketone bodies constitute an important source of oxidative fuel for neurones in an environment of glucose restriction. Importantly, the lactate shuttle between astrocytes and neurones is dependent on glycogenolysis and glycolysis, resulting from the fact that the astrocytic filopodia responsible for lactate release are too narrow to accommodate mitochondria. The entry into the CNS of ketone bodies and fatty acids, as a result of nutritional ketosis, has effects on the astrocytic glutamate–glutamine cycle, glutamate synthase activity, and on the function of vesicular glutamate transporters, EAAT, Na+, K+-ATPase, Kir4.1, aquaporin-4, Cx34 and KATP channels, as well as on astrogliosis. These mechanisms are detailed and it is suggested that they would tend to mitigate the changes seen in many neurodegenerative and neuroprogressive disorders. Hence, it is hypothesized that nutritional ketosis may have therapeutic applications in such disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | | |
Collapse
|
33
|
Braidy N, Liu Y. NAD+ therapy in age-related degenerative disorders: A benefit/risk analysis. Exp Gerontol 2020; 132:110831. [PMID: 31917996 DOI: 10.1016/j.exger.2020.110831] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential pyridine nucleotide that is present in all living cells. NAD+ acts as an important cofactor and substrate for a multitude of biological processes including energy production, DNA repair, gene expression, calcium-dependent secondary messenger signalling and immunoregulatory roles. The de novo synthesis of NAD+ is primarily dependent on the kynurenine pathway (KP), although NAD+ can also be recycled from nicotinic acid (NA), nicotinamide (NAM) and nicotinamide riboside (NR). NAD+ levels have been reported to decline during ageing and age-related diseases. Recent studies have shown that raising intracellular NAD+ levels represents a promising therapeutic strategy for age-associated degenerative diseases in general and to extend lifespan in small animal models. A systematic review of the literature available on Medline, Embase and Pubmed was undertaken to evaluate the potential health and/or longevity benefits due to increasing NAD+ levels. A total of 1545 articles were identified and 147 articles (113 preclinical and 34 clinical) met criteria for inclusion. Most studies indicated that the NAD+ precursors NAM, NR, nicotinamide mononucleotide (NMN), and to a lesser extent NAD+ and NADH had a favourable outcome on several age-related disorders associated with the accumulation of chronic oxidative stress, inflammation and impaired mitochondrial function. While these compounds presented with a limited acute toxicity profile, evidence is still quite limited and long-term human clinical trials are still nascent in the current literature. Potential risks in raising NAD+ levels in various clinical disorders using NAD+ precursors include the accumulation of putative toxic metabolites, tumorigenesis and promotion of cellular senescence. Therefore, NAD+ metabolism represents a promising target and further studies are needed to recapitulate the preclinical benefits in human clinical trials.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia.
| | - Yue Liu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
34
|
Morris G, Puri BK, Walker AJ, Berk M, Walder K, Bortolasci CC, Marx W, Carvalho AF, Maes M. The compensatory antioxidant response system with a focus on neuroprogressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109708. [PMID: 31351160 DOI: 10.1016/j.pnpbp.2019.109708] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
Major antioxidant responses to increased levels of inflammatory, oxidative and nitrosative stress (ONS) are detailed. In response to increasing levels of nitric oxide, S-nitrosylation of cysteine thiol groups leads to post-transcriptional modification of many cellular proteins and thereby regulates their activity and allows cellular adaptation to increased levels of ONS. S-nitrosylation inhibits the function of nuclear factor kappa-light-chain-enhancer of activated B cells, toll-like receptor-mediated signalling and the activity of several mitogen-activated protein kinases, while activating nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2 or NFE2L2); in turn, the redox-regulated activation of Nrf2 leads to increased levels and/or activity of key enzymes and transporter systems involved in the glutathione system. The Nrf2/Kelch-like ECH-associated protein-1 axis is associated with upregulation of NAD(P)H:quinone oxidoreductase 1, which in turn has anti-inflammatory effects. Increased Nrf2 transcriptional activity also leads to activation of haem oxygenase-1, which is associated with upregulation of bilirubin, biliverdin and biliverdin reductase as well as increased carbon monoxide signalling, anti-inflammatory and antioxidant activity. Associated transcriptional responses, which may be mediated by retrograde signalling owing to elevated hydrogen peroxide, include the unfolded protein response (UPR), mitohormesis and the mitochondrial UPR; the UPR also results from increasing levels of mitochondrial and cytosolic reactive oxygen species and reactive nitrogen species leading to nitrosylation, glutathionylation, oxidation and nitration of crucial cysteine and tyrosine causing protein misfolding and the development of endoplasmic reticulum stress. It is shown how these mechanisms co-operate in forming a co-ordinated rapid and prolonged compensatory antioxidant response system.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Adam J Walker
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry, The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ken Walder
- CMMR Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Chiara C Bortolasci
- CMMR Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Wolfgang Marx
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Andre F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.
| | - Michael Maes
- IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
35
|
Lee DC, Vali K, Baldwin SR, Divino JN, Feliciano JL, Fequiere JR, Fernandez MA, Frageau JC, Longo FK, Madhoun SS, Mingione V P, O’Toole TR, Ruiz MG, Tanner GR. Dietary Supplementation With the Ketogenic Diet Metabolite Beta-Hydroxybutyrate Ameliorates Post-TBI Aggression in Young-Adult Male Drosophila. Front Neurosci 2019; 13:1140. [PMID: 31736687 PMCID: PMC6833482 DOI: 10.3389/fnins.2019.01140] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI), caused by repeated concussive head trauma can induce chronic traumatic encephalopathy (CTE), a neurodegenerative disease featuring behavioral symptoms ranging from cognitive deficits to elevated aggression. In a Drosophila model, we used a high-impact trauma device (Katzenberger et al., 2013, 2015) to induce TBI-like symptoms and to study post-TBI behavioral outcomes. Following TBI, aggression in banged male flies was significantly elevated as compared with that in unbanged flies. These increases in aggressive behavior were not the result of basal motility changes, as measured by a negative geotaxis assay. In addition, the increase in post-TBI aggression appeared to be specific to concussive trauma: neither cold exposure nor electric shock-two alternate types of trauma-significantly elevated aggressive behavior in male-male pairs. Various forms of dietary therapy, especially the high-fat, low-carbohydrate ketogenic diet (KD), have recently been explored for a wide variety of neuropathies. We thus hypothesized that putatively neuroprotective dietary interventions might be able to suppress post-traumatic elevations in aggressive behavior in animals subjected to head-trauma-inducing strikes, or "bangs". We supplemented a normal high-carbohydrate Drosophila diet with the KD metabolite beta-hydroxybutyrate (β-HB)-a ketone body (KB). Banged flies raised on a KB-supplemented diet exhibited a marked reduction in aggression, whereas aggression in unbanged flies was equivalent whether dieted with KB supplements or not. Pharmacological blockade of the ATP-sensitive potassium (KATP) channel abrogated KB effects reducing post-TBI aggression while pharmacological activation mimicked them, suggesting a mechanism by which KBs act in this model. KBs did not significantly extend lifespan in banged flies, but markedly extended lifespan in unbanged flies. We have thus developed a functional model for the study of post-TBI elevations of aggression. Further, we conclude that dietary interventions may be a fruitful avenue for further exploration of treatments for TBI- and CTE-related cognitive-behavioral symptoms.
Collapse
Affiliation(s)
- Derek C. Lee
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- The Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, United States
| | - Krishna Vali
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- The Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, United States
| | - Shane R. Baldwin
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Jeffrey N. Divino
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Justin L. Feliciano
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Jesus R. Fequiere
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Mirella A. Fernandez
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - James C. Frageau
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- The Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, United States
| | - Frank K. Longo
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Salaheddine S. Madhoun
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Pasquale Mingione V
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Timothy R. O’Toole
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- The Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, United States
| | - Maria G. Ruiz
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Geoffrey R. Tanner
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- The Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, United States
| |
Collapse
|
36
|
Alves-Fernandes DK, Jasiulionis MG. The Role of SIRT1 on DNA Damage Response and Epigenetic Alterations in Cancer. Int J Mol Sci 2019; 20:E3153. [PMID: 31261609 PMCID: PMC6651129 DOI: 10.3390/ijms20133153] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022] Open
Abstract
Sirtuin-1 (SIRT1) is a class-III histone deacetylase (HDAC), an NAD+-dependent enzyme deeply involved in gene regulation, genome stability maintenance, apoptosis, autophagy, senescence, proliferation, aging, and tumorigenesis. It also has a key role in the epigenetic regulation of tissue homeostasis and many diseases by deacetylating both histone and non-histone targets. Different studies have shown ambiguous implications of SIRT1 as both a tumor suppressor and tumor promoter. However, this contradictory role seems to be determined by the cell type and SIRT1 localization. SIRT1 upregulation has already been demonstrated in some cancer cells, such as acute myeloid leukemia (AML) and primary colon, prostate, melanoma, and non-melanoma skin cancers, while SIRT1 downregulation was described in breast cancer and hepatic cell carcinomas. Even though new functions of SIRT1 have been characterized, the underlying mechanisms that define its precise role on DNA damage and repair and their contribution to cancer development remains underexplored. Here, we discuss the recent findings on the interplay among SIRT1, oxidative stress, and DNA repair machinery and its impact on normal and cancer cells.
Collapse
Affiliation(s)
| | - Miriam Galvonas Jasiulionis
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil.
| |
Collapse
|
37
|
Veech RL, Todd King M, Pawlosky R, Kashiwaya Y, Bradshaw PC, Curtis W. The "great" controlling nucleotide coenzymes. IUBMB Life 2019; 71:565-579. [PMID: 30624851 PMCID: PMC6850382 DOI: 10.1002/iub.1997] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
Nucleotide coenzymes dot the map of metabolic pathways providing energy to drive the reactions of the pathway and play an important role in regulating and controlling energy metabolism through their shared potential energy, which is widely unobserved due to the paradox that the energy in the coenzyme pools cannot be determined from the concentration of the coenzyme couples. The potential energy of the nucleotide couples in the mitochondria or the cytoplasm is expressed in the enzyme reactions in which they take part. The energy in these couples, [NAD+]/[NADH], [NADP+]/[NADPH], [acetyl CoA]/[CoA], and [ATP]/[ADP]x[Pi], regulates energy metabolism. The energy contained in the couples can be altered by suppling energy equivalents in the form of ketones, such as, D-β-hydroxybutyrate to overcome insulin resistance, to restore antioxidants capacity, to form potential treatments for Alzheimer's and Parkinson's diseases, to enhance life span, and to increase physiological performance. © 2019 IUBMB Life, 71(5):565-579, 2019.
Collapse
Affiliation(s)
- Richard L Veech
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | - Michael Todd King
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | - Robert Pawlosky
- Laboratory of Metabolic Control, NIAAA, NIH, Rockville, MD, 20852, USA
| | | | - Patrick C Bradshaw
- Department of Biomedical Sciences, East Tennessee State University College of Medicine, Johnson City, TN, USA
| | - William Curtis
- Department of Biomedical Sciences, East Tennessee State University College of Medicine, Johnson City, TN, USA
| |
Collapse
|