1
|
Ye Z, Liu R, Wang H, Zuo A, Jin C, Wang N, Sun H, Feng L, Yang H. Neuroprotective potential for mitigating ischemia-reperfusion-induced damage. Neural Regen Res 2025; 20:2199-2217. [PMID: 39104164 PMCID: PMC11759025 DOI: 10.4103/nrr.nrr-d-23-01985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/09/2024] [Accepted: 06/22/2024] [Indexed: 08/07/2024] Open
Abstract
Reperfusion following cerebral ischemia causes both structural and functional damage to brain tissue and could aggravate a patient's condition; this phenomenon is known as cerebral ischemia-reperfusion injury. Current studies have elucidated the neuroprotective role of the sirtuin protein family (Sirtuins) in modulating cerebral ischemia-reperfusion injury. However, the potential of utilizing it as a novel intervention target to influence the prognosis of cerebral ischemia-reperfusion injury requires additional exploration. In this review, the origin and research progress of Sirtuins are summarized, suggesting the involvement of Sirtuins in diverse mechanisms that affect cerebral ischemia-reperfusion injury, including inflammation, oxidative stress, blood-brain barrier damage, apoptosis, pyroptosis, and autophagy. The therapeutic avenues related to Sirtuins that may improve the prognosis of cerebral ischemia-reperfusion injury were also investigated by modulating Sirtuins expression and affecting representative pathways, such as nuclear factor-kappa B signaling, oxidative stress mediated by adenosine monophosphate-activated protein kinase, and the forkhead box O. This review also summarizes the potential of endogenous substances, such as RNA and hormones, drugs, dietary supplements, and emerging therapies that regulate Sirtuins expression. This review also reveals that regulating Sirtuins mitigates cerebral ischemia-reperfusion injury when combined with other risk factors. While Sirtuins show promise as a potential target for the treatment of cerebral ischemia-reperfusion injury, most recent studies are based on rodent models with circadian rhythms that are distinct from those of humans, potentially influencing the efficacy of Sirtuins-targeting drug therapies. Overall, this review provides new insights into the role of Sirtuins in the pathology and treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Zi Ye
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Runqing Liu
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hangxing Wang
- Division of Infectious Diseases, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aizhen Zuo
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Cen Jin
- School of Medical Imaging, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Nan Wang
- Division of Gastroenterology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huiqi Sun
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Luqian Feng
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hua Yang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
2
|
Vahid ZF, Eskandani M, Dadashi H, Vandghanooni S, Rashidi MR. Recent advances in potential enzymes and their therapeutic inhibitors for the treatment of Alzheimer's disease. Heliyon 2024; 10:e40756. [PMID: 39717593 PMCID: PMC11664286 DOI: 10.1016/j.heliyon.2024.e40756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024] Open
Abstract
Alzheimer's disease (AD), a chronic neurodegenerative disease, is clinically characterized by loss of memory and learning ability among other neurological deficits. Amyloid plaques, hyperphosphorylated tau protein, and neurofibrillary tangles involve in AD etiology. Meanwhile, enzymes and their inhibitors have become the focus of research in AD treatment. In this review, the molecular mechanisms involved in the pathogenesis of AD were overviewed and various enzymes such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), β-secretase, γ-secretase, monoamine oxidase (MAO), and receptor of advanced glycation end products (RAGE) were highlighted as potential targets for AD treatment. Several hybrid molecules with essential substructures derived from various chemotypes have demonstrated desired pharmacological activity. It is envisioned that the development of new drugs that inhibit enzymes involved in AD is a future trend in the management of the disease.
Collapse
Affiliation(s)
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Dadashi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Medicinal Chemistry Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Bonomi RE, Riordan W, Gelovani JG. The Structures, Functions, and Roles of Class III HDACs (Sirtuins) in Neuropsychiatric Diseases. Cells 2024; 13:1644. [PMID: 39404407 PMCID: PMC11476333 DOI: 10.3390/cells13191644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Over the past two decades, epigenetic regulation has become a rapidly growing and influential field in biology and medicine. One key mechanism involves the acetylation and deacetylation of lysine residues on histone core proteins and other critical proteins that regulate gene expression and cellular signaling. Although histone deacetylases (HDACs) have received significant attention, the roles of individual HDAC isoforms in the pathogenesis of psychiatric diseases still require further research. This is particularly true with regard to the sirtuins, class III HDACs. Sirtuins have unique functional activity and significant roles in normal neurophysiology, as well as in the mechanisms of addiction, mood disorders, and other neuropsychiatric abnormalities. This review aims to elucidate the differences in catalytic structure and function of the seven sirtuins as they relate to psychiatry.
Collapse
Affiliation(s)
- Robin E. Bonomi
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA;
| | - William Riordan
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA;
| | - Juri G. Gelovani
- College of Medicine and Health Sciences, Office of the Provost, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Radiology, Division of Nuclear Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
4
|
Patel D, Soni R, Shah J. Decoding the Role of Nuclear Sirtuins in Parkinson's Pathogenesis. ACS Chem Neurosci 2024. [PMID: 39331405 DOI: 10.1021/acschemneuro.4c00507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
Parkinson's disease (PD) is the second most prevailing degenerative disease that deals with dopaminergic neuronal loss and deficiency of dopamine in SNpc and striatum. Manifestations primarily include motor symptoms like tremor, rigidity, and akinesia/dyskinesia along with some nonmotor symptoms like GI and olfactory dysfunction. α-Synuclein pathogenesis is the major cause behind progression of PD; however there are many underlying molecular mechanisms behind the pathophysiology of PD. Sirtuins are small molecular deacetylases that have an imperative role in pathology of such neurodegenerative disorders like PD. Sirtuins are majorly classified according to their location; nuclear (SIRT1,7,6), mitochondrial sirtuins (SIRT3-5), and cytosolic (SIRT2). These actively take part in pathological development and possess independent actions. In this review, the role of nuclear sirtuins is individualistically explored for better understanding of PD pathology and development of advanced therapeutics targeting sirtuins.
Collapse
Affiliation(s)
- Dishank Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Ritu Soni
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| |
Collapse
|
5
|
Yuan T, Kumar S, Skinner ME, Victor-Joseph R, Abuaita M, Keijer J, Zhang J, Kunkel TJ, Liu Y, Petrunak EM, Saunders TL, Lieberman AP, Stuckey JA, Neamati N, Al-Murshedi F, Alfadhel M, Spelbrink JN, Rodenburg R, de Boer VC, Lombard DB. Human SIRT5 variants with reduced stability and activity do not cause neuropathology in mice. iScience 2024; 27:109991. [PMID: 38846003 PMCID: PMC11154205 DOI: 10.1016/j.isci.2024.109991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/06/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
SIRT5 is a sirtuin deacylase that removes negatively charged lysine modifications, in the mitochondrial matrix and elsewhere in the cell. In benign cells and mouse models, under basal conditions, the phenotypes of SIRT5 deficiency are quite subtle. Here, we identify two homozygous SIRT5 variants in patients suspected to have mitochondrial disease. Both variants, P114T and L128V, are associated with reduced SIRT5 protein stability and impaired biochemical activity, with no evidence of neomorphic or dominant negative properties. The crystal structure of the P114T enzyme was solved and shows only subtle deviations from wild-type. Via CRISPR-Cas9, we generated a mouse model that recapitulates the human P114T mutation; homozygotes show reduced SIRT5 levels and activity, but no obvious metabolic abnormalities, neuropathology, or other gross phenotypes. We conclude that these human SIRT5 variants most likely represent severe hypomorphs, but are likely not by themselves the primary pathogenic cause of the neuropathology observed in the patients.
Collapse
Affiliation(s)
- Taolin Yuan
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, the Netherlands
| | - Surinder Kumar
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mary E. Skinner
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan Victor-Joseph
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Majd Abuaita
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, the Netherlands
| | - Jessica Zhang
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Thaddeus J. Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanghan Liu
- Department of Medicinal Chemistry, College of Pharmacy and Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elyse M. Petrunak
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas L. Saunders
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Jeanne A. Stuckey
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy and Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fathiya Al-Murshedi
- Genetic and Developmental Medicine Clinic, Department of Genetics, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Majid Alfadhel
- Medical Genomic Research Department, King Abdullah International Medical Research Center(KAIMRC), King Saud Bin Abdulaziz University for Health Sciences(KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
- Genetics and Precision Medicine Department (GPM), King Abdullah Specialized Children’s Hospital (KASCH), King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
| | - Johannes N. Spelbrink
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Richard Rodenburg
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vincent C.J. de Boer
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, the Netherlands
| | - David B. Lombard
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
- Miami VA Healthcare System, Miami, FL 33125, USA
| |
Collapse
|
6
|
Yuan T, Kumar S, Skinner M, Victor-Joseph R, Abuaita M, Keijer J, Zhang J, Kunkel TJ, Liu Y, Petrunak EM, Saunders TL, Lieberman AP, Stuckey JA, Neamati N, Al-Murshedi F, Alfadhel M, Spelbrink JN, Rodenburg R, de Boer VCJ, Lombard DB. SIRT5 variants from patients with mitochondrial disease are associated with reduced SIRT5 stability and activity, but not with neuropathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570371. [PMID: 38105987 PMCID: PMC10723467 DOI: 10.1101/2023.12.06.570371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
SIRT5 is a sirtuin deacylase that represents the major activity responsible for removal of negatively-charged lysine modifications, in the mitochondrial matrix and elsewhere in the cell. In benign cells and mouse models, under basal non-stressed conditions, the phenotypes of SIRT5 deficiency are generally quite subtle. Here, we identify two homozygous SIRT5 variants in human patients suffering from severe mitochondrial disease. Both variants, P114T and L128V, are associated with reduced SIRT5 protein stability and impaired biochemical activity, with no evidence of neomorphic or dominant negative properties. The crystal structure of the P114T enzyme was solved and shows only subtle deviations from wild-type. Via CRISPR-Cas9, we generate a mouse model that recapitulates the human P114T mutation; homozygotes show reduced SIRT5 levels and activity, but no obvious metabolic abnormalities, neuropathology or other gross evidence of severe disease. We conclude that these human SIRT5 variants most likely represent severe hypomorphs, and are likely not the primary pathogenic cause of the neuropathology observed in the patients.
Collapse
Affiliation(s)
- Taolin Yuan
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, The Netherlands
| | - Surinder Kumar
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami FL 33136
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Mary Skinner
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | | | - Majd Abuaita
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, The Netherlands
| | - Jessica Zhang
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami FL 33136
| | | | - Yanghan Liu
- Department of Medicinal Chemistry, College of Pharmacy and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109
| | - Elyse M. Petrunak
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Thomas L. Saunders
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | | | - Jeanne A. Stuckey
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109
| | - Fathiya Al-Murshedi
- Genetic and Developmental Medicine Clinic, Department of Genetics, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Majid Alfadhel
- Medical Genomic Research Department, King Abdullah International Medical Research Center(KAIMRC), King Saud Bin Abdulaziz University for Health Sciences(KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
- Genetics and Precision Medicine department (GPM), King Abdullah Specialized Children’s Hospital (KASCH), King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
| | - Johannes N. Spelbrink
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Richard Rodenburg
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vincent C. J. de Boer
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, The Netherlands
| | - David B. Lombard
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami FL 33136
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
- Miami VA Healthcare System, Miami FL 33125
| |
Collapse
|
7
|
Oh HSH, Rutledge J, Nachun D, Pálovics R, Abiose O, Moran-Losada P, Channappa D, Urey DY, Kim K, Sung YJ, Wang L, Timsina J, Western D, Liu M, Kohlfeld P, Budde J, Wilson EN, Guen Y, Maurer TM, Haney M, Yang AC, He Z, Greicius MD, Andreasson KI, Sathyan S, Weiss EF, Milman S, Barzilai N, Cruchaga C, Wagner AD, Mormino E, Lehallier B, Henderson VW, Longo FM, Montgomery SB, Wyss-Coray T. Organ aging signatures in the plasma proteome track health and disease. Nature 2023; 624:164-172. [PMID: 38057571 PMCID: PMC10700136 DOI: 10.1038/s41586-023-06802-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
Animal studies show aging varies between individuals as well as between organs within an individual1-4, but whether this is true in humans and its effect on age-related diseases is unknown. We utilized levels of human blood plasma proteins originating from specific organs to measure organ-specific aging differences in living individuals. Using machine learning models, we analysed aging in 11 major organs and estimated organ age reproducibly in five independent cohorts encompassing 5,676 adults across the human lifespan. We discovered nearly 20% of the population show strongly accelerated age in one organ and 1.7% are multi-organ agers. Accelerated organ aging confers 20-50% higher mortality risk, and organ-specific diseases relate to faster aging of those organs. We find individuals with accelerated heart aging have a 250% increased heart failure risk and accelerated brain and vascular aging predict Alzheimer's disease (AD) progression independently from and as strongly as plasma pTau-181 (ref. 5), the current best blood-based biomarker for AD. Our models link vascular calcification, extracellular matrix alterations and synaptic protein shedding to early cognitive decline. We introduce a simple and interpretable method to study organ aging using plasma proteomics data, predicting diseases and aging effects.
Collapse
Affiliation(s)
- Hamilton Se-Hwee Oh
- Graduate Program in Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jarod Rutledge
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Graduate Program in Genetics, Stanford University, Stanford, CA, USA
| | - Daniel Nachun
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Róbert Pálovics
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Olamide Abiose
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia Moran-Losada
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Divya Channappa
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Deniz Yagmur Urey
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University School of Engineering, Stanford, CA, USA
| | - Kate Kim
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Dan Western
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Pat Kohlfeld
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John Budde
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Edward N Wilson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yann Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Taylor M Maurer
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Haney
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew C Yang
- Departments of Neurology and Anatomy, University of California San Francisco, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Bakar Aging Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrin I Andreasson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Sanish Sathyan
- Departments of Medicine and Genetics, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
| | - Erica F Weiss
- Department of Neurology, Montefiore Medical Center, New York, NY, USA
| | - Sofiya Milman
- Departments of Medicine and Genetics, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
| | - Nir Barzilai
- Departments of Medicine and Genetics, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Anthony D Wagner
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Elizabeth Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Victor W Henderson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | - Frank M Longo
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephen B Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
8
|
Scieszka D, Bolt AM, McCormick MA, Brigman JL, Campen MJ. Aging, longevity, and the role of environmental stressors: a focus on wildfire smoke and air quality. FRONTIERS IN TOXICOLOGY 2023; 5:1267667. [PMID: 37900096 PMCID: PMC10600394 DOI: 10.3389/ftox.2023.1267667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Aging is a complex biological process involving multiple interacting mechanisms and is being increasingly linked to environmental exposures such as wildfire smoke. In this review, we detail the hallmarks of aging, emphasizing the role of telomere attrition, cellular senescence, epigenetic alterations, proteostasis, genomic instability, and mitochondrial dysfunction, while also exploring integrative hallmarks - altered intercellular communication and stem cell exhaustion. Within each hallmark of aging, our review explores how environmental disasters like wildfires, and their resultant inhaled toxicants, interact with these aging mechanisms. The intersection between aging and environmental exposures, especially high-concentration insults from wildfires, remains under-studied. Preliminary evidence, from our group and others, suggests that inhaled wildfire smoke can accelerate markers of neurological aging and reduce learning capabilities. This is likely mediated by the augmentation of circulatory factors that compromise vascular and blood-brain barrier integrity, induce chronic neuroinflammation, and promote age-associated proteinopathy-related outcomes. Moreover, wildfire smoke may induce a reduced metabolic, senescent cellular phenotype. Future interventions could potentially leverage combined anti-inflammatory and NAD + boosting compounds to counter these effects. This review underscores the critical need to study the intricate interplay between environmental factors and the biological mechanisms of aging to pave the way for effective interventions.
Collapse
Affiliation(s)
- David Scieszka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Alicia M. Bolt
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Mark A. McCormick
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jonathan L. Brigman
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
9
|
Liu Y, Wang L, Yang G, Chi X, Liang X, Zhang Y. Sirtuins: Promising Therapeutic Targets to Treat Ischemic Stroke. Biomolecules 2023; 13:1210. [PMID: 37627275 PMCID: PMC10452362 DOI: 10.3390/biom13081210] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Stroke is a major cause of mortality and disability globally, with ischemic stroke (IS) accounting for over 80% of all stroke cases. The pathological process of IS involves numerous signal molecules, among which are the highly conserved nicotinamide adenine dinucleotide (NAD+)-dependent enzymes known as sirtuins (SIRTs). SIRTs modulate various biological processes, including cell differentiation, energy metabolism, DNA repair, inflammation, and oxidative stress. Importantly, several studies have reported a correlation between SIRTs and IS. This review introduces the general aspects of SIRTs, including their distribution, subcellular location, enzyme activity, and substrate. We also discuss their regulatory roles and potential mechanisms in IS. Finally, we describe the current therapeutic methods based on SIRTs, such as pharmacotherapy, non-pharmacological therapeutic/rehabilitative interventions, epigenetic regulators, potential molecules, and stem cell-derived exosome therapy. The data collected in this study will potentially contribute to both clinical and fundamental research on SIRTs, geared towards developing effective therapeutic candidates for future treatment of IS.
Collapse
Affiliation(s)
- Yue Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Y.L.); (L.W.); (X.C.)
| | - Liuding Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Y.L.); (L.W.); (X.C.)
| | - Guang Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China;
| | - Xiansu Chi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Y.L.); (L.W.); (X.C.)
| | - Xiao Liang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Y.L.); (L.W.); (X.C.)
| | - Yunling Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Y.L.); (L.W.); (X.C.)
| |
Collapse
|
10
|
Paula Ceballos M, Darío Quiroga A, Palma NF. Role of sirtuins in hepatocellular carcinoma progression and multidrug resistance: Mechanistical and pharmacological perspectives. Biochem Pharmacol 2023; 212:115573. [PMID: 37127248 DOI: 10.1016/j.bcp.2023.115573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of death from cancer worldwide. Therapeutic strategies are still challenging due to the high relapse rate after surgery and multidrug resistance (MDR). It is essential to better understand the mechanisms for HCC progression and MDR for the development of new therapeutic strategies. Mammalian sirtuins (SIRTs), a family of seven members, are related to tumor progression, MDR and prognosis and were proposed as potential prognostic markers, as well as therapeutic targets for treating cancer. SIRT1 is the most studied member and is overexpressed in HCC, playing an oncogenic role and predicting poor prognosis. Several manuscripts describe the role of SIRTs2-7 in HCC; most of them report an oncogenic role for SIRT2 and -7 and a suppressive role for SIRT3 and -4. The scenario is more confusing for SIRT5 and -6, since information is contradictory and scarce. For SIRT1 many inhibitors are available and they seem to hold therapeutic promise in HCC. For the other members the development of specific modulators has just started. This review is aimed to describe the features of SIRTs1-7 in HCC, and the role they play in the onset and progression of the disease. Also, when possible, we will depict the information related to the SIRTs modulators that have been tested in HCC and their possible implication in MDR. With this, we hope to clarify the role of each member in HCC and to shed some light on the most successful strategies to overcome MDR.
Collapse
Affiliation(s)
- María Paula Ceballos
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, UNR, Suipacha 70 (S2002LRL), Rosario, Argentina.
| | - Ariel Darío Quiroga
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, UNR, Suipacha 70 (S2002LRL), Rosario, Argentina; Área Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Suipachs 570 (S2002LRL), Rosario, Argentina; Centro de Altos Estudios en Ciencias Humanas y de la Salud (CAECIHS) Sede Regional Rosario, Universidad Abierta Interamericana, Av. Pellegrini 1618 (S2000BUG), Rosario, Argentina
| | - Nicolás Francisco Palma
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, UNR, Suipacha 70 (S2002LRL), Rosario, Argentina; Área Morfología, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, Suipachs 570 (S2002LRL), Rosario, Argentina
| |
Collapse
|
11
|
Xing J, Chen K, Gao S, Pousse M, Ying Y, Wang B, Chen L, Wang C, Wang L, Hu W, Lu Y, Gilson E, Ye J. Protein phosphatase 2A activators reverse age-related behavioral changes by targeting neural cell senescence. Aging Cell 2023; 22:e13780. [PMID: 36644807 PMCID: PMC10014060 DOI: 10.1111/acel.13780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/04/2022] [Accepted: 12/19/2022] [Indexed: 01/17/2023] Open
Abstract
The contribution of cellular senescence to the behavioral changes observed in the elderly remains elusive. Here, we observed that aging is associated with a decline in protein phosphatase 2A (PP2A) activity in the brains of zebrafish and mice. Moreover, drugs activating PP2A reversed age-related behavioral changes. We developed a transgenic zebrafish model to decrease PP2A activity in the brain through knockout of the ppp2r2c gene encoding a regulatory subunit of PP2A. Mutant fish exhibited the behavioral phenotype observed in old animals and premature accumulation of neural cells positive for markers of cellular senescence, including senescence-associated β-galactosidase, elevated levels cdkn2a/b, cdkn1a, senescence-associated secretory phenotype gene expression, and an increased level of DNA damage signaling. The behavioral and cell senescence phenotypes were reversed in mutant fish through treatment with the senolytic ABT263 or diverse PP2A activators as well as through cdkn1a or tp53 gene ablation. Senomorphic function of PP2A activators was demonstrated in mouse primary neural cells with downregulated Ppp2r2c. We conclude that PP2A reduction leads to neural cell senescence thereby contributing to age-related behavioral changes and that PP2A activators have senotherapeutic properties against deleterious behavioral effects of brain aging.
Collapse
Affiliation(s)
- Jun Xing
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| | - Kehua Chen
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| | - Shuaiyun Gao
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| | - Mélanie Pousse
- International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China.,IRCAN, Côte d'Azur University, CNRS, Inserm, Nice, France
| | - Yilin Ying
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China.,IRCAN, Côte d'Azur University, CNRS, Inserm, Nice, France
| | - Bo Wang
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| | - Lianxiang Chen
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| | - Cuicui Wang
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| | - Lei Wang
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiguo Hu
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Lu
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| | - Eric Gilson
- International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China.,IRCAN, Côte d'Azur University, CNRS, Inserm, Nice, France.,Department of Genetics, CHU, Nice, France
| | - Jing Ye
- Geriatric Department, Geriatric Medical Center, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,International Laboratory in Cancer, Aging and Hematology, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur University, Shanghai, China
| |
Collapse
|
12
|
Varela-López A, Ramírez-Tortosa CL, Ramos-Pleguezuelos FM, Márquez-Lobo B, Battino M, Quiles JL. Differences reported in the lifespan and aging of male Wistar rats maintained on diets containing fat with different fatty acid profiles (virgin olive, sunflower or fish oils) are not reflected by histopathological lesions found at death in central nervous and endocrine systems. Food Chem Toxicol 2022; 168:113357. [PMID: 35985366 DOI: 10.1016/j.fct.2022.113357] [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: 05/20/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022]
Abstract
The present study was designed to examine if dietary fat sources that have shown differences in lifespan and if some aging-related aspects can modulate the range of histopathologic changes in central nervous and endocrine systems that occur during the lifespan of Wistar rats. Moreover, it was attempted to gain insight into the relationship between longevity and the development of the different pathological changes, as well as possible interaction with diet. In order to achieve this, male Wistar rats were randomly assigned to three experimental groups fed semisynthetic and isoenergetic diets from weaning until death with different dietary fat sources, namely virgin olive, sunflower, or fish oil. An individual follow-up until death of each animal was performed. Incidence, severity, and burden of specific or group (i.e., neoplastic or non-neoplastic proliferative and non-proliferative) of lesions was calculated along with individual's disease and individual organ lesion burden. Most of the histopathological lesions found have been described in previous studies. Neoplasms, and in particular pituitary adenomas followed by brain tumors, were the most prevalent lesions found in the rats and the main cause of death involving both systems. Incidence of brain lesions was associated with age-at-death. Assayed dietary fats did not present differential effects on pathological changes occurring in endocrine and central nervous systems throughout rat lifespan.
Collapse
Affiliation(s)
- Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "Jose Mataix Verdú," Biomedical Research Center, University of Granada, Armilla, Granada, Spain.
| | | | | | | | - Maurizio Battino
- Department of Clinical Sciences, Polytechnic University of Marche, 60131, Ancona, Italy; International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang, 212013, China
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "Jose Mataix Verdú," Biomedical Research Center, University of Granada, Armilla, Granada, Spain; Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres, 21, 39011, Santander, Spain.
| |
Collapse
|
13
|
Dada O, Jeremian R, Dai N, Strauss J, Zai C, Graff A, De Luca V. Investigation of accelerated epigenetic aging in individuals experiencing suicidal ideation. Schizophr Res 2022; 243:307-309. [PMID: 33041163 DOI: 10.1016/j.schres.2020.09.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/17/2020] [Accepted: 09/26/2020] [Indexed: 10/23/2022]
Affiliation(s)
| | - Richie Jeremian
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - Nasia Dai
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - John Strauss
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - Clement Zai
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - Ariel Graff
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | | |
Collapse
|
14
|
Stone WS, Phillips MR, Yang LH, Kegeles LS, Susser ES, Lieberman JA. Neurodegenerative model of schizophrenia: Growing evidence to support a revisit. Schizophr Res 2022; 243:154-162. [PMID: 35344853 PMCID: PMC9189010 DOI: 10.1016/j.schres.2022.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/21/2022]
Abstract
Multidimensional progressive declines in the absence of standard biomarkers for neurodegeneration are observed commonly in the development of schizophrenia, and are accepted as consistent with neurodevelopmental etiological hypotheses to explain the origins of the disorder. Far less accepted is the possibility that neurodegenerative processes are involved as well, or even that key dimensions of function, such as cognition and aspects of biological integrity, such as white matter function, decline in chronic schizophrenia beyond levels associated with normal aging. We propose that recent research germane to these issues warrants a current look at the question of neurodegeneration. We propose the view that a neurodegenerative hypothesis provides a better explanation of some features of chronic schizophrenia, including accelerated aging, than is provided by neurodevelopmental hypotheses. Moreover, we suggest that neurodevelopmental influences in early life, including those that may extend to later life, do not preclude the development of neurodegenerative processes in later life, including some declines in cognitive and biological integrity. We evaluate these views by integrating recent findings in representative domains such as cognition and white and gray matter integrity with results from studies on accelerated aging, together with functional implications of neurodegeneration for our understanding of chronic schizophrenia.
Collapse
Affiliation(s)
- William S. Stone
- Harvard Medical School Department of Psychiatry at Beth Israel Deaconess Medical Center, Boston, Massachusetts,Corresponding Author: William S. Stone, Ph.D., Massachusetts Mental Health Center, 75 Fenwood Road, Boston, Massachusetts, USA,
| | - Michael R. Phillips
- Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, China,Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | - Lawrence H. Yang
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York,New York University College of Global Public Health, New York, New York
| | - Lawrence S. Kegeles
- Department of Psychiatry, Columbia University, New York, New York,New York State Psychiatric Institute, New York, New York
| | - Ezra S. Susser
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | | |
Collapse
|
15
|
Mahjabeen I, Rizwan M, Fareen G, Waqar Ahmed M, Farooq Khan A, Akhtar Kayani M. Mitochondrial sirtuins genetic variations and gastric cancer risk: Evidence from retrospective observational study. Gene 2022; 807:145951. [PMID: 34500051 DOI: 10.1016/j.gene.2021.145951] [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] [Received: 03/10/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022]
Abstract
AIMS The purpose of the present study was to analyze the role of selected polymorphisms of SIRT3 and SIRT5 in gastric carcinogenesis. METHODS For this study, 500 blood samples of GC patients and 500 blood samples of healthy individuals were collected. Six selected polymorphisms of mitochondrial sirtuins were analyzed for analysis using Tetra-Arms PCR followed by DNA sequencing. RESULTS Mutant allele frequencies of selected polymorphisms [rs3782116 (p < 0.0001), rs6598072 (p < 0.0001) and rs11246020 (p < 0.0001), rs938222 (p = 0.0136), rs3757261 (p = 0.0005) and rs2841511 (p = 0.0015)] were observed significant higher in GC patients vs controls. Haplotype analysis was performed, and 51 haplotypes were generated using haploview software. Among these haplotypes, eleven haplotypes were found associated with a significantly increased risk of GC. Furthermore, SNP-SNP interaction showed a significant correlation between studied SNPs and GC risk. Kaplan Meier analysis showed that mutant allele frequencies of selected polymorphisms are linked with a significant decrease in survival of GC patients CONCLUSIONS: It can be concluded that selected SNPs may be associated with enhanced risk of GC and hence can be potential prognostic markers for prognosis and predisposition of GC.
Collapse
Affiliation(s)
- Ishrat Mahjabeen
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Muhammad Rizwan
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Gul Fareen
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Malik Waqar Ahmed
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan; Pakistan Institute of Rehabilitation Sciences (PIRS), Isra University Islamabad Campus, Islamabad, Pakistan
| | | | - Mahmood Akhtar Kayani
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan.
| |
Collapse
|
16
|
Brain age and Alzheimer's-like atrophy are domain-specific predictors of cognitive impairment in Parkinson's disease. Neurobiol Aging 2021; 109:31-42. [PMID: 34649002 DOI: 10.1016/j.neurobiolaging.2021.08.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 11/20/2022]
Abstract
Recently, it was shown that patients with Parkinson's disease (PD) who exhibit an "Alzheimer's disease (AD)-like" pattern of brain atrophy are at greater risk for future cognitive decline. This study aimed to investigate whether this association is domain-specific and whether atrophy associated with brain aging also relates to cognitive impairment in PD. SPARE-AD, an MRI index capturing AD-like atrophy, and atrophy-based estimates of brain age were computed from longitudinal structural imaging data of 178 PD patients and 84 healthy subjects from the LANDSCAPE cohort. All patients underwent an extensive neuropsychological test battery. Patients diagnosed with mild cognitive impairment or dementia were found to have higher SPARE-AD scores as compared to patients with normal cognition and healthy controls. All patient groups showed increased brain age. SPARE-AD predicted impairment in memory, language and executive functions, whereas advanced brain age was associated with deficits in attention and working memory. Data suggest that SPARE-AD and brain age are differentially related to domain-specific cognitive decline in PD. The underlying pathomechanisms remain to be determined.
Collapse
|
17
|
Satapathy MK, Yen TL, Jan JS, Tang RD, Wang JY, Taliyan R, Yang CH. Solid Lipid Nanoparticles (SLNs): An Advanced Drug Delivery System Targeting Brain through BBB. Pharmaceutics 2021; 13:1183. [PMID: 34452143 PMCID: PMC8402065 DOI: 10.3390/pharmaceutics13081183] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) plays a vital role in the protection and maintenance of homeostasis in the brain. In this way, it is an interesting target as an interface for various types of drug delivery, specifically in the context of the treatment of several neuropathological conditions where the therapeutic agents cannot cross the BBB. Drug toxicity and on-target specificity are among some of the limitations associated with current neurotherapeutics. In recent years, advances in nanodrug delivery have enabled the carrier system containing the active therapeutic drug to target the signaling pathways and pathophysiology that are closely linked to central nervous system (CNS) disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), brain tumor, epilepsy, ischemic stroke, and neurodegeneration. At present, among the nano formulations, solid lipid nanoparticles (SLNs) have emerged as a putative drug carrier system that can deliver the active therapeutics (drug-loaded SLNs) across the BBB at the target site of the brain, offering a novel approach with controlled drug delivery, longer circulation time, target specificity, and higher efficacy, and more importantly, reducing toxicity in a biomimetic way. This paper highlights the synthesis and application of SLNs as a novel nontoxic formulation strategy to carry CNS drugs across the BBB to improve the use of therapeutics agents in treating major neurological disorders in future clinics.
Collapse
Affiliation(s)
- Mantosh Kumar Satapathy
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
| | - Ting-Lin Yen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
- Department of Medical Research, Cathay General Hospital, Taipei 22174, Taiwan
| | - Jing-Shiun Jan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
| | - Ruei-Dun Tang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan;
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan;
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan
- Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Rajeev Taliyan
- Department of Pharmacy, Neuropsychopharmacology Division, Birla Institute of Technology and Science, Pilani 333031, India;
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
- Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
18
|
Lin CW, Chang LC, Ma T, Oh H, French B, Puralewski R, Mathews F, Fang Y, Lewis DA, Kennedy JL, Mueller D, Marshe VS, Jaffe A, Chen Q, Ursini G, Weinberger D, Newman AB, Lenze EJ, Nikolova YS, Tseng GC, Sibille E. Older molecular brain age in severe mental illness. Mol Psychiatry 2021; 26:3646-3656. [PMID: 32632206 PMCID: PMC7785531 DOI: 10.1038/s41380-020-0834-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 06/03/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022]
Abstract
Psychiatric disorders are associated with accelerated aging and enhanced risk for neurodegenerative disorders. Brain aging is associated with molecular, cellular, and structural changes that are robust on the group level, yet show substantial inter-individual variability. Here we assessed deviations in gene expression from normal age-dependent trajectories, and tested their validity as predictors of risk for major mental illnesses and neurodegenerative disorders. We performed large-scale gene expression and genotype analyses in postmortem samples of two frontal cortical brain regions from 214 control subjects aged 20-90 years. Individual estimates of "molecular age" were derived from age-dependent genes, identified by robust regression analysis. Deviation from chronological age was defined as "delta age". Genetic variants associated with deviations from normal gene expression patterns were identified by expression quantitative trait loci (cis-eQTL) of age-dependent genes or genome-wide association study (GWAS) on delta age, combined into distinct polygenic risk scores (PRScis-eQTL and PRSGWAS), and tested for predicting brain disorders or pathology in independent postmortem expression datasets and clinical cohorts. In these validation datasets, molecular ages, defined by 68 and 76 age-related genes for two brain regions respectively, were positively correlated with chronological ages (r = 0.88/0.91), elevated in bipolar disorder (BP) and schizophrenia (SCZ), and unchanged in major depressive disorder (MDD). Exploratory analyses in independent clinical datasets show that PRSs were associated with SCZ and MDD diagnostics, and with cognition in SCZ and pathology in Alzheimer's disease (AD). These results suggest that older molecular brain aging is a common feature of severe mental illnesses and neurodegeneration.
Collapse
Affiliation(s)
- Chien-Wei Lin
- Department of Biostatistics, Graduate school of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Lun-Ching Chang
- Department of Biostatistics, Graduate school of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Mathematical Sciences, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Tianzhou Ma
- Department of Biostatistics, Graduate school of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- Department of Epidemiology and Biostatistics, University of Maryland, College Park, MD, 20742, USA
| | - Hyunjung Oh
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15312, USA
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, M5T1R8, ON, Canada
| | - Beverly French
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15312, USA
| | - Rachel Puralewski
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15312, USA
| | - Fasil Mathews
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15312, USA
| | - Yusi Fang
- Department of Biostatistics, Graduate school of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15312, USA
| | - James L Kennedy
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, M5T1R8, ON, Canada
| | - Daniel Mueller
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, M5T1R8, ON, Canada
| | - Victoria S Marshe
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, M5T1R8, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Andrew Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Gianluca Ursini
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Daniel Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Anne B Newman
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric J Lenze
- Department of Psychiatry, Washington University, St. Louis, MO, 63130, USA
| | - Yuliya S Nikolova
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, M5T1R8, ON, Canada
| | - George C Tseng
- Department of Biostatistics, Graduate school of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Etienne Sibille
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15312, USA.
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, M5T1R8, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5T1R8, ON, Canada.
| |
Collapse
|
19
|
Dada O, Adanty C, Dai N, Jeremian R, Alli S, Gerretsen P, Graff A, Strauss J, De Luca V. Biological aging in schizophrenia and psychosis severity: DNA methylation analysis. Psychiatry Res 2021; 296:113646. [PMID: 33444986 DOI: 10.1016/j.psychres.2020.113646] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/10/2020] [Indexed: 01/08/2023]
Abstract
The physiological changes associated with normal aging are known to occur earlier in individuals with schizophrenia (SCZ). One of the phenomena linked with normal aging is the change in patterns of epigenetic modifications. We recruited 138 individuals with SCZ spectrum disorders and extracted DNA from white blood cells. The combinations of pre-selected DNA methylation sites were utilized to estimate the 'methylation age' (DNAm age) and evaluate evidence of epigenetic age acceleration. We investigated the correlation between the epigenetic age acceleration measures and psychosis severity; furthermore, we estimated blood cell counts based on DNA methylation levels. The extrinsic epigenetic age acceleration showed a significant correlation with the Brief Psychiatric Rating Scale (BPRS) disorganization subscale(r=0.222, p=0.039).Both Horvath age acceleration and Hannum age acceleration showed a significant correlation (r=0.221, p=0.029; r=0.242, p=0.017 respectively) with the Symptom Checklist 90 (SCL-90) psychotic domain. Overall, this study shows some evidence of epigenetic age acceleration associated with psychosis severity using two different algorithms for DNAm age analysis.
Collapse
Affiliation(s)
- Oluwagbenga Dada
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Christopher Adanty
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Nasia Dai
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Richie Jeremian
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Sauliha Alli
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Philip Gerretsen
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Ariel Graff
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - John Strauss
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Vincenzo De Luca
- CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
20
|
Goetzman ES, Bharathi SS, Zhang Y, Zhao XJ, Dobrowolski SF, Peasley K, Sims-Lucas S, Monga SP. Impaired mitochondrial medium-chain fatty acid oxidation drives periportal macrovesicular steatosis in sirtuin-5 knockout mice. Sci Rep 2020; 10:18367. [PMID: 33110171 PMCID: PMC7591893 DOI: 10.1038/s41598-020-75615-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
Medium-chain triglycerides (MCT), containing C8-C12 fatty acids, are used to treat several pediatric disorders and are widely consumed as a nutritional supplement. Here, we investigated the role of the sirtuin deacylase Sirt5 in MCT metabolism by feeding Sirt5 knockout mice (Sirt5KO) high-fat diets containing either C8/C10 fatty acids or coconut oil, which is rich in C12, for five weeks. Coconut oil, but not C8/C10 feeding, induced periportal macrovesicular steatosis in Sirt5KO mice. 14C-C12 degradation was significantly reduced in Sirt5KO liver. This decrease was localized to the mitochondrial β-oxidation pathway, as Sirt5KO mice exhibited no change in peroxisomal C12 β-oxidation. Endoplasmic reticulum ω-oxidation, a minor fatty acid degradation pathway known to be stimulated by C12 accumulation, was increased in Sirt5KO liver. Mice lacking another mitochondrial C12 oxidation enzyme, long-chain acyl-CoA dehydrogenase (LCAD), also developed periportal macrovesicular steatosis when fed coconut oil, confirming that defective mitochondrial C12 oxidation is sufficient to induce the steatosis phenotype. Sirt5KO liver exhibited normal LCAD activity but reduced mitochondrial acyl-CoA synthetase activity with C12. These studies reveal a role for Sirt5 in regulating the hepatic response to MCT and may shed light into the pathogenesis of periportal steatosis, a hallmark of human pediatric non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Eric S Goetzman
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Sivakama S Bharathi
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yuxun Zhang
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xue-Jun Zhao
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven F Dobrowolski
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kevin Peasley
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sunder Sims-Lucas
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Satdarshan P Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Kanzato N, Nakachi K, Naka T, Mochizuki S, Miyamae Y, Okada Y. Parkinson's disease therapy with Istradefylline and blood biomarkers of epigenetics. ACTA ACUST UNITED AC 2020. [DOI: 10.1111/ncn3.12415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Naomi Kanzato
- Department of Neurology Okinawa Prefectural Southern Medical Center & Children's Medical Center Okinawa Japan
| | - Kou Nakachi
- Department of Neurology Okinawa Prefectural Southern Medical Center & Children's Medical Center Okinawa Japan
| | - Taro Naka
- Department of Neurology Okinawa Prefectural Southern Medical Center & Children's Medical Center Okinawa Japan
| | - Satsuki Mochizuki
- Department of Surgery National Defense Medical College Saitama Japan
| | - Yuka Miyamae
- Department of Pathophysiology for Locomotive and Neoplastic Diseases Juntendo University Graduate School of Medicine Tokyo Japan
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases Juntendo University Graduate School of Medicine Tokyo Japan
| |
Collapse
|
22
|
Shukla R, Oh H, Sibille E. Molecular and Cellular Evidence for Age by Disease Interactions: Updates and Path Forward. Am J Geriatr Psychiatry 2020; 28:237-247. [PMID: 31285153 DOI: 10.1016/j.jagp.2019.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/14/2019] [Accepted: 06/01/2019] [Indexed: 12/31/2022]
Abstract
Characterization of age-associated gene expression changes shows that the brain engages a specific set of genes and biologic pathways along a continuous life-long trajectory and that these genes and pathways overlap with those associated with brain-related disorders. Based on this correlative observation, we have suggested a model of age-by-disease interaction by which brain ageing promotes biologic changes associated with diseases and where deviations from expected age-related trajectories, due to biologic and environmental factors, contribute to defining disease risk or resiliency. In this review, we first evaluate various biomarkers that can be used to study age-by-disease interactions and then focus on transcriptome analysis (i.e., the set of all expressed genes) as a useful tool to explore this interaction. Using the specific example of brain-derived neurotrophic factor and brain-derived neurotrophic factor-associated genes, we then describe molecular events and mechanisms potentially contributing to age-by-disease interactions. Finally, we suggest that long-term biologic adaptations within distinct cellular components of cortical microcircuits, as determined by transcriptome analysis, may integrate and mediate the effects of ageing and diseases. Moving forward, we suggest that analysis of transcriptome similarities between ageing and small molecule-induced system perturbations may lead to novel therapeutics discovery.
Collapse
Affiliation(s)
- Rammohan Shukla
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Hyunjung Oh
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
23
|
Samal J, Rebelo AL, Pandit A. A window into the brain: Tools to assess pre-clinical efficacy of biomaterials-based therapies on central nervous system disorders. Adv Drug Deliv Rev 2019; 148:68-145. [PMID: 30710594 DOI: 10.1016/j.addr.2019.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/04/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic conveyance into the brain is a cardinal requirement for treatment of diverse central nervous system (CNS) disorders and associated pathophysiology. Effectual shielding of the brain by the blood-brain barrier (BBB) sieves out major proportion of therapeutics with the exception of small lipophilic molecules. Various nano-delivery systems (NDS) provide an effective solution around this obstacle owing to their small size and targeting properties. To date, these systems have been used for several pre-clinical disease models including glioma, neurodegenerative diseases and psychotic disorders. An efficacy screen for these systems involves a test battery designed to probe into the multiple facets of therapeutic delivery. Despite their wide application in redressing various disease targets, the efficacy evaluation strategies for all can be broadly grouped into four modalities, namely: histological, bio-imaging, molecular and behavioural. This review presents a comprehensive insight into all of these modalities along with their strengths and weaknesses as well as perspectives on an ideal design for a panel of tests to screen brain nano-delivery systems.
Collapse
Affiliation(s)
- Juhi Samal
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ana Lucia Rebelo
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
24
|
Foolad F, Khodagholi F, Javan M. Sirtuins in Multiple Sclerosis: The crossroad of neurodegeneration, autoimmunity and metabolism. Mult Scler Relat Disord 2019; 34:47-58. [PMID: 31228716 DOI: 10.1016/j.msard.2019.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/26/2019] [Accepted: 06/07/2019] [Indexed: 12/17/2022]
Abstract
Multiple Sclerosis (MS) is a challenging and disabling condition particularly in the secondary progressive (SP) phase of this disease. The available treatments cannot ameliorate or stop disease progression in this phase, and there is an urgent need to focus on effective therapies and the molecular pathways involved SPMS. Given the significant impact of neurodegeneration, autoimmunity and metabolic alterations in MS, focusing on the molecules that target these different pathways could help in finding new treatments. Sirtuins (SIRTs) are NAD+ dependent epigenetic and metabolic regulators, which have critical roles in the physiology of central nervous system, immune system and metabolism. Based on these facts, SIRTs are crucial candidates of therapeutic targets in MS and collecting the information related to MS disease for each SIRT individually is noteworthy and highlights the lack of investigation in each part. In this review we summarized the role of different sirtuins as key regulator in neurodegeneration, autoimmunity and metabolism pathways. We also clarify the rationale behind selecting SIRTs as therapeutic targets in MS disease by collecting the researches showing alteration of these proteins in human samples of MS patients and animal model of MS, and also the improvement of modeled animals after SIRT-directed treatments.
Collapse
Affiliation(s)
- Forough Foolad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
25
|
Glorioso CA, Pfenning AR, Lee SS, Bennett DA, Sibille EL, Kellis M, Guarente LP. Rate of brain aging and APOE ε4 are synergistic risk factors for Alzheimer's disease. Life Sci Alliance 2019; 2:2/3/e201900303. [PMID: 31133613 PMCID: PMC6537750 DOI: 10.26508/lsa.201900303] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
This study describes a gauge for brain aging predictive of cognitive decline and Alzheimer's disease synergistic with the APOE ε4 allele. Advanced age and the APOE ε4 allele are the two biggest risk factors for Alzheimer’s disease (AD) and declining cognitive function. We describe a universal gauge to measure molecular brain age using transcriptome analysis of four human postmortem cohorts (n = 673, ages 25–97) free of neurological disease. In a fifth cohort of older subjects with or without neurological disease (n = 438, ages 67–108), we show that subjects with brains deviating in the older direction from what would be expected based on chronological age show an increase in AD, Parkinson’s disease, and cognitive decline. Strikingly, a younger molecular age (−5 yr than chronological age) protects against AD even in the presence of APOE ε4. An established DNA methylation gauge for age correlates well with the transcriptome gauge for determination of molecular age and assigning deviations from the expected. Our results suggest that rapid brain aging and APOE ε4 are synergistic risk factors, and interventions that slow aging may substantially reduce risk of neurological disease and decline even in the presence of APOE ε4.
Collapse
Affiliation(s)
- Christin A Glorioso
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA .,Paul F. Glenn Center for Biology of Aging Research at Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andreas R Pfenning
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Sam S Lee
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Paul F. Glenn Center for Biology of Aging Research at Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Etienne L Sibille
- Department of Psychiatry and of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.,Campbell Family Mental Health Research Institute, Toronto, Ontario, Canada
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA.,The Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Leonard P Guarente
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA .,Paul F. Glenn Center for Biology of Aging Research at Massachusetts Institute of Technology, Cambridge, MA, USA.,The Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
26
|
Chen L, Wang H, Gao F, Zhang J, Zhang Y, Ma R, Pang S, Cui Y, Yang J, Yan B. Functional genetic variants in the SIRT5 gene promoter in acute myocardial infarction. Gene 2018; 675:233-239. [DOI: 10.1016/j.gene.2018.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/14/2018] [Accepted: 07/03/2018] [Indexed: 01/03/2023]
|
27
|
Intracellular and Intercellular Signalling Mechanisms following DNA Damage Are Modulated By PINK1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1391387. [PMID: 30116473 PMCID: PMC6079383 DOI: 10.1155/2018/1391387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/04/2018] [Accepted: 05/20/2018] [Indexed: 02/06/2023]
Abstract
Impaired mitochondrial function and accumulation of DNA damage have been recognized as hallmarks of age-related diseases. Mitochondrial dysfunction initiates protective signalling mechanisms coordinated at nuclear level particularly by modulating transcription of stress signalling factors. In turn, cellular response to DNA lesions comprises a series of interconnected complex protective pathways, which require the energetic and metabolic support of the mitochondria. These are involved in intracellular as well as in extracellular signalling of damage. Here, we have initiated a study that addresses how mitochondria-nucleus communication may occur in conditions of combined mitochondrial dysfunction and genotoxic stress and what are the consequences of this interaction on the cell system. In this work, we used cells deficient for PINK1, a mitochondrial kinase involved in mitochondrial quality control whose loss of function leads to the accumulation of dysfunctional mitochondria, challenged with inducers of DNA damage, namely, ionizing radiation and the radiomimetic bleomycin. Combined stress at the level of mitochondria and the nucleus impairs both mitochondrial and nuclear functions. Our findings revealed exacerbated sensibility to genotoxic stress in PINK1-deficient cells. The same cells showed an impaired induction of bystander phenomena following stress insults. However, these cells responded adaptively when a challenge dose was applied subsequently to a low-dose treatment to the cells. The data demonstrates that PINK1 modulates intracellular and intercellular signalling pathways, particularly adaptive responses and transmission of bystander signalling, two facets of the cell-protective mechanisms against detrimental agents.
Collapse
|
28
|
McKinney BC, Lin H, Ding Y, Lewis DA, Sweet RA. DNA methylation age is not accelerated in brain or blood of subjects with schizophrenia. Schizophr Res 2018; 196:39-44. [PMID: 28988914 PMCID: PMC5886835 DOI: 10.1016/j.schres.2017.09.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/18/2017] [Accepted: 09/21/2017] [Indexed: 01/01/2023]
Abstract
Individuals with schizophrenia (SZ) exhibit multiple premature age-related phenotypes and die ~20years prematurely. The accelerated aging hypothesis of SZ has been advanced to explain these observations, it posits that SZ-associated factors accelerate the progressive biological changes associated with normal aging. Testing the hypothesis has been limited by the absence of robust, meaningful, and multi-tissue measures of biological age. Recently, a method was described in which DNA methylation (DNAm) levels at 353 genomic sites are used to produce "DNAm age", an estimate of biological age with advantages over existing measures. We used this method and 3 publicly-available DNAm datasets, 1 from brain and 2 from blood, to test the hypothesis. The brain dataset was composed of data from the dorsolateral prefrontal cortex of 232 non-psychiatric control (NPC) and 195 SZ subjects. Blood dataset #1 was composed of data from whole blood of 304 NPC and 332 SZ subjects, and blood dataset #2 was composed of data from whole blood of 405 NPC and 260 SZ subjects. DNAm age and chronological age correlated strongly (r=0.92-0.95, p<0.0001) in both NPC and SZ subjects in all 3 datasets. DNAm age acceleration did not differ between NPC and SZ subjects in the brain dataset (t=0.52, p=0.60), blood dataset #1 (t=1.51, p=0.13), or blood dataset #2 (t=0.93, p=0.35). Consistent with our previous findings from a smaller study of postmortem brains, our findings suggest there is no acceleration of brain or blood aging in SZ and, thus, do not support the accelerated aging hypothesis of SZ.
Collapse
Affiliation(s)
| | - Huang Lin
- University of Pittsburgh, Department of Biostatistics, Pittsburgh, PA
| | - Ying Ding
- University of Pittsburgh, Department of Biostatistics, Pittsburgh, PA
| | - David A. Lewis
- University of Pittsburgh, Department of Psychiatry, Pittsburgh, PA
| | - Robert A. Sweet
- University of Pittsburgh, Department of Psychiatry, Pittsburgh, PA,University of Pittsburgh, Department of Neurology, Pittsburgh, PA,VISN4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA
| |
Collapse
|
29
|
Kumar S, Lombard DB. Functions of the sirtuin deacylase SIRT5 in normal physiology and pathobiology. Crit Rev Biochem Mol Biol 2018; 53:311-334. [PMID: 29637793 DOI: 10.1080/10409238.2018.1458071] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sirtuins are NAD+-dependent protein deacylases/ADP-ribosyltransferases that have emerged as candidate targets for new therapeutics to treat metabolic disorders and other diseases, including cancer. The sirtuin SIRT5 resides primarily in the mitochondrial matrix and catalyzes the removal of negatively charged lysine acyl modifications; succinyl, malonyl, and glutaryl groups. Evidence has now accumulated to document the roles of SIRT5 as a significant regulator of cellular homeostasis, in a context- and cell-type specific manner, as has been observed previously for other sirtuin family members. SIRT5 regulates protein substrates involved in glycolysis, the TCA cycle, fatty acid oxidation, electron transport chain, ketone body formation, nitrogenous waste management, and ROS detoxification, among other processes. SIRT5 plays pivotal roles in cardiac physiology and stress responses and is involved in the regulation of numerous aspects of myocardial energy metabolism. SIRT5 is implicated in neoplasia, as both a tumor promoter and suppressor in a context-specific manner, and may serve a protective function in the setting of neurodegenerative disorders. Here, we review the current understanding of functional impacts of SIRT5 on its metabolic targets, and its molecular functions in both normal and pathological conditions. Finally, we will discuss the potential utility of SIRT5 as a drug target and also summarize the current status, progress, and challenges in developing small molecule compounds to modulate SIRT5 activity with high potency and specificity.
Collapse
Affiliation(s)
- Surinder Kumar
- a Department of Pathology , University of Michigan , Ann Arbor , MI , USA
| | - David B Lombard
- a Department of Pathology , University of Michigan , Ann Arbor , MI , USA.,b Institute of Gerontology , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
30
|
Abstract
SIGNIFICANCE Developing evidence in the literature suggests that sirtuin 5 (SIRT5) may be involved in metabolic reprogramming, an emerging hallmark of cancer by which neoplastic cells reconfigure their metabolism to support the anabolic demands of rapid cell division. SIRT5 is one of the seven members of the nicotinamide adenine dinucleotide-dependent sirtuin family of lysine deacetylases. It removes succinyl, malonyl, and glutaryl groups from protein targets within the mitochondrial matrix and other subcellular compartments. SIRT5 substrates include a number of proteins integral to metabolism. Recent Advances: New work has begun to elucidate the roles of SIRT5 in glycolysis, tricarboxylic acid cycle, fatty acid oxidation, nitrogen metabolism, pentose phosphate pathway, antioxidant defense, and apoptosis. CRITICAL ISSUES In this study, we summarize biological functions of SIRT5 reported in normal tissues and in cancer and discuss potential mechanisms whereby SIRT5 may impact tumorigenesis, particularly focusing on its reported roles in metabolic reprogramming. Finally, we review current efforts to target SIRT5 pharmacologically. FUTURE DIRECTIONS The biological significance of SIRT5 has been elucidated in the context of only an extremely small fraction of its targets and interactors. There is no doubt that further studies in this area will provide a wealth of insights into functions of SIRT5 and its targets in normal and neoplastic cells. Antioxid. Redox Signal. 28, 677-690.
Collapse
Affiliation(s)
| | - Angela H. Guo
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - David B. Lombard
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
31
|
Uddin R, Singh SM. Gene Network Construction from Microarray Data Identifies a Key Network Module and Several Candidate Hub Genes in Age-Associated Spatial Learning Impairment. Front Syst Neurosci 2017; 11:75. [PMID: 29066959 PMCID: PMC5641338 DOI: 10.3389/fnsys.2017.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/22/2017] [Indexed: 01/06/2023] Open
Abstract
As humans age many suffer from a decrease in normal brain functions including spatial learning impairments. This study aimed to better understand the molecular mechanisms in age-associated spatial learning impairment (ASLI). We used a mathematical modeling approach implemented in Weighted Gene Co-expression Network Analysis (WGCNA) to create and compare gene network models of young (learning unimpaired) and aged (predominantly learning impaired) brains from a set of exploratory datasets in rats in the context of ASLI. The major goal was to overcome some of the limitations previously observed in the traditional meta- and pathway analysis using these data, and identify novel ASLI related genes and their networks based on co-expression relationship of genes. This analysis identified a set of network modules in the young, each of which is highly enriched with genes functioning in broad but distinct GO functional categories or biological pathways. Interestingly, the analysis pointed to a single module that was highly enriched with genes functioning in “learning and memory” related functions and pathways. Subsequent differential network analysis of this “learning and memory” module in the aged (predominantly learning impaired) rats compared to the young learning unimpaired rats allowed us to identify a set of novel ASLI candidate hub genes. Some of these genes show significant repeatability in networks generated from independent young and aged validation datasets. These hub genes are highly co-expressed with other genes in the network, which not only show differential expression but also differential co-expression and differential connectivity across age and learning impairment. The known function of these hub genes indicate that they play key roles in critical pathways, including kinase and phosphatase signaling, in functions related to various ion channels, and in maintaining neuronal integrity relating to synaptic plasticity and memory formation. Taken together, they provide a new insight and generate new hypotheses into the molecular mechanisms responsible for age associated learning impairment, including spatial learning.
Collapse
Affiliation(s)
- Raihan Uddin
- Department of Biology, University of Western Ontario, London, ON, Canada
| | - Shiva M Singh
- Department of Biology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
32
|
Mitochondrial Respiration in Intact Peripheral Blood Mononuclear Cells and Sirtuin 3 Activity in Patients with Movement Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9703574. [PMID: 29081897 PMCID: PMC5610844 DOI: 10.1155/2017/9703574] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/01/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Mitochondrial dysfunction is considered a unifying pathophysiological explanation for movement disorders. Sirtuin 3 (SIRT3) exhibits deacetylase activity and antioxidant properties. The aim of the study was to analyze the mitochondrial respiration in peripheral blood mononuclear cells (PBMCs) and the SIRT3 activity in patients with movement disorders. METHODS Mitochondrial respiration was analyzed in intact PBMCs using the ROUTINE, LEAK, electron transfer system (ETS), and residual oxygen consumption (ROX) protocol by means of high-resolution respirometry. The SIRT3 expression and PBMC activity were measured using fluorometry. Ultrasound measurements of the echogenicity of the substantia nigra and the diameter of the 3rd ventricle were also performed. RESULTS Patients with movement disorders exhibited a lower ROUTINE respiration than controls (P = 0.0237). Reduced oxygen fluxes in the LEAK (P = 0.033) and ROX (P = 0.0486) states were observed in patients with movement disorders compared with controls. Decreased ROUTINE respiration (P = 0.007) and oxygen flux in the LEAK state (P = 0.0203) were observed in patients with PD with substantia nigra hyperechogenicity compared with controls. Decreased SIRT 3 deacetylase activity was found in patients with movement disorders. CONCLUSION Impaired mitochondrial respiration in intact PBMCs was associated with inhibited SIRT3 activity and neurodegeneration measures evaluated using ultrasound in patients with PD.
Collapse
|
33
|
Collier TJ, Kanaan NM, Kordower JH. Aging and Parkinson's disease: Different sides of the same coin? Mov Disord 2017; 32:983-990. [PMID: 28520211 PMCID: PMC5844262 DOI: 10.1002/mds.27037] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/10/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023] Open
Abstract
Despite abundant epidemiological evidence in support of aging as the primary risk factor for PD, biological correlates of a connection have been elusive. In this article, we address the following question: does aging represent biology accurately characterized as pre-PD? We present evidence from our work on midbrain dopamine neurons of aging nonhuman primates that demonstrates that markers of known correlates of dopamine neuron degeneration in PD, including impaired proteasome/lysosome function, oxidative/nitrative damage, and inflammation, all increase with advancing age and are exaggerated in the ventral tier substantia nigra dopamine neurons most vulnerable to degeneration in PD. Our findings support the view that aging-related changes in the dopamine system approach the biological threshold for parkinsonism, actively producing a vulnerable pre-parkinsonian state. © 2017 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Timothy J. Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Mercy Health Hauenstein Neuroscience Center, Grand Rapids, Michigan, USA
| | - Nicholas M. Kanaan
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan, USA
- Mercy Health Hauenstein Neuroscience Center, Grand Rapids, Michigan, USA
| | - Jeffrey H. Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
34
|
McKinney BC, Lin H, Ding Y, Lewis DA, Sweet RA. DNA methylation evidence against the accelerated aging hypothesis of schizophrenia. NPJ SCHIZOPHRENIA 2017; 3:13. [PMID: 28560259 PMCID: PMC5441537 DOI: 10.1038/s41537-017-0017-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/20/2017] [Accepted: 03/06/2017] [Indexed: 01/19/2023]
Abstract
The accelerated aging hypothesis of schizophrenia posits that physiological changes throughout the body that are associated with normal aging occur at an earlier age in individuals with schizophrenia. Testing this hypothesis has been limited by problems measuring biological age. Recently, a method using DNA methylation levels at 353 genomic sites to produce "DNA methylation age", an estimate of tissue biological age, was described and validated. We used this method to test the hypothesis in the postmortem superior temporal gyrus of 22 non-psychiatric control and 22 schizophrenia subjects. DNA methylation age correlated with chronological age in both non-psychiatric control (r = 0.95, p < 0.0001) and schizophrenia subjects (r = 0.96, p < 0.0001). Age acceleration did not differ between non-psychiatric control and schizophrenia subjects (t = 1.27, p = 0.21). Our findings suggest there is no acceleration of brain aging in schizophrenia. Larger studies using samples from multiple brain regions and homogenous cell populations will be necessary to confirm these findings.
Collapse
Affiliation(s)
| | - Huang Lin
- Departments of Biostatistics, University of Pittsburgh, Pittsburgh, PA USA
| | - Ying Ding
- Departments of Biostatistics, University of Pittsburgh, Pittsburgh, PA USA
| | - David A. Lewis
- Departments of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Robert A. Sweet
- Departments of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
- Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| |
Collapse
|
35
|
Douillard-Guilloux G, Lewis D, Seney ML, Sibille E. Decrease in somatostatin-positive cell density in the amygdala of females with major depression. Depress Anxiety 2017; 34:68-78. [PMID: 27557481 PMCID: PMC5222785 DOI: 10.1002/da.22549] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 07/29/2016] [Accepted: 07/30/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Somatostatin (SST) is a neuropeptide expressed in a subtype of gamma-aminobutyric acid (GABA) interneurons that target the dendrites of pyramidal neurons. We previously reported reduced levels of SST gene and protein expression in the postmortem amygdala of subjects with major depressive disorder (MDD). This reduction was specific to female subjects with MDD. METHODS Here, we used in situ hybridization to examine the regional and cellular patterns of reductions in SST expression in a cohort of female MDD subjects with known SST deficits in the amygdala (N = 10/group). RESULTS We report a significant reduction in the density of SST-labeled neurons in the lateral, basolateral, and basomedial nuclei of the amygdala of MDD subjects compared to controls. SST mRNA levels per neuron did not differ between MDD and control subjects in the lateral or basolateral nuclei, but were lower in the basomedial nucleus. There was no difference in cross-sectional density of total cells. CONCLUSIONS In summary, we report an MDD-related reduction in the density of detectable SST-positive neurons across several nuclei in the amygdala, with a reduction in SST mRNA per cell restricted to the basomedial nucleus. In the absence of changes in total cell density, these results suggest the possibility of a change in SST cell phenotype rather than cell death in the amygdala of female MDD subjects.
Collapse
Affiliation(s)
| | - David Lewis
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marianne L. Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA,Corresponding authors: Marianne L. Seney, 450 Technology Drive, Bridgeside Point II Room 226, Pittsburgh, PA 15219, Phone: 412-624-3072; Fax: 412-624-5280, ; Etienne Sibille, 250 College Street, Toronto, ON M5T 1R8, Phone: 416-535-8501, ext 36571,
| | - Etienne Sibille
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA,Campbell Family Mental Health Research Institute of CAMH; Departments of Psychiatry, and of Pharmacology and Toxicology, University of Toronto, Toronto, CA,Corresponding authors: Marianne L. Seney, 450 Technology Drive, Bridgeside Point II Room 226, Pittsburgh, PA 15219, Phone: 412-624-3072; Fax: 412-624-5280, ; Etienne Sibille, 250 College Street, Toronto, ON M5T 1R8, Phone: 416-535-8501, ext 36571,
| |
Collapse
|
36
|
Sirtuin 5: a review of structure, known inhibitors and clues for developing new inhibitors. SCIENCE CHINA-LIFE SCIENCES 2016; 60:249-256. [DOI: 10.1007/s11427-016-0060-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
|
37
|
Application of Targeted Mass Spectrometry for the Quantification of Sirtuins in the Central Nervous System. Sci Rep 2016; 6:35391. [PMID: 27762282 PMCID: PMC5071856 DOI: 10.1038/srep35391] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 09/28/2016] [Indexed: 02/05/2023] Open
Abstract
Sirtuin proteins have a variety of intracellular targets, thereby regulating multiple biological pathways including neurodegeneration. However, relatively little is currently known about the role or expression of the 7 mammalian sirtuins in the central nervous system. Western blotting, PCR and ELISA are the main techniques currently used to measure sirtuin levels. To achieve sufficient sensitivity and selectivity in a multiplex-format, a targeted mass spectrometric assay was developed and validated for the quantification of all seven mammalian sirtuins (SIRT1-7). Quantification of all peptides was by multiple reaction monitoring (MRM) using three mass transitions per protein-specific peptide, two specific peptides for each sirtuin and a stable isotope labelled internal standard. The assay was applied to a variety of samples including cultured brain cells, mammalian brain tissue, CSF and plasma. All sirtuin peptides were detected in the human brain, with SIRT2 being the most abundant. Sirtuins were also detected in human CSF and plasma, and guinea pig and mouse tissues. In conclusion, we have successfully applied MRM mass spectrometry for the detection and quantification of sirtuin proteins in the central nervous system, paving the way for more quantitative and functional studies.
Collapse
|
38
|
Khojah SM, Payne AP, McGuinness D, Shiels PG. Segmental Aging Underlies the Development of a Parkinson Phenotype in the AS/AGU Rat. Cells 2016; 5:E38. [PMID: 27763519 PMCID: PMC5187522 DOI: 10.3390/cells5040038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 09/30/2016] [Accepted: 10/01/2016] [Indexed: 12/15/2022] Open
Abstract
There is a paucity of information on the molecular biology of aging processes in the brain. We have used biomarkers of aging (SA β-Gal, p16Ink4a, Sirt5, Sirt6, and Sirt7) to demonstrate the presence of an accelerated aging phenotype across different brain regions in the AS/AGU rat, a spontaneous Parkinsonian mutant of PKCγ derived from a parental AS strain. P16INK4a expression was significantly higher in AS/AGU animals compared to age-matched AS controls (p < 0.001) and displayed segmental expression across various brain regions. The age-related expression of sirtuins similarly showed differences between strains and between brain regions. Our data clearly show segmental aging processes within the rat brain, and that these are accelerated in the AS/AGU mutant. The accelerated aging, Parkinsonian phenotype, and disruption to dopamine signalling in the basal ganglia in AS/AGU rats, suggests that this rat strain represents a useful model for studies of development and progression of Parkinson's disease in the context of biological aging and may offer unique mechanistic insights into the biology of aging.
Collapse
Affiliation(s)
- Sohair M Khojah
- School of Life Sciences, Pharmacology Research Theme, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Anthony P Payne
- School of Life Sciences, Pharmacology Research Theme, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Dagmara McGuinness
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| | - Paul G Shiels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| |
Collapse
|
39
|
Gertz M, Steegborn C. Using mitochondrial sirtuins as drug targets: disease implications and available compounds. Cell Mol Life Sci 2016; 73:2871-96. [PMID: 27007507 PMCID: PMC11108305 DOI: 10.1007/s00018-016-2180-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/15/2016] [Accepted: 03/11/2016] [Indexed: 02/06/2023]
Abstract
Sirtuins are an evolutionary conserved family of NAD(+)-dependent protein lysine deacylases. Mammals have seven Sirtuin isoforms, Sirt1-7. They contribute to regulation of metabolism, stress responses, and aging processes, and are considered therapeutic targets for metabolic and aging-related diseases. While initial studies were focused on Sirt1 and 2, recent progress on the mitochondrial Sirtuins Sirt3, 4, and 5 has stimulated research and drug development for these isoforms. Here we review the roles of Sirtuins in regulating mitochondrial functions, with a focus on the mitochondrially located isoforms, and on their contributions to disease pathologies. We further summarize the compounds available for modulating the activity of these Sirtuins, again with a focus on mitochondrial isoforms, and we describe recent results important for the further improvement of compounds. This overview illustrates the potential of mitochondrial Sirtuins as drug targets and summarizes the status, progress, and challenges in developing small molecule compounds modulating their activity.
Collapse
Affiliation(s)
- Melanie Gertz
- Department of Biochemistry, University of Bayreuth, Universitätsstr. 30, 95447, Bayreuth, Germany
- Bayer Pharma AG, Apratherweg 18a, 42096, Wuppertal, Germany
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, Universitätsstr. 30, 95447, Bayreuth, Germany.
| |
Collapse
|
40
|
Zhou L, Wang F, Sun R, Chen X, Zhang M, Xu Q, Wang Y, Wang S, Xiong Y, Guan KL, Yang P, Yu H, Ye D. SIRT5 promotes IDH2 desuccinylation and G6PD deglutarylation to enhance cellular antioxidant defense. EMBO Rep 2016; 17:811-22. [PMID: 27113762 DOI: 10.15252/embr.201541643] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/08/2016] [Indexed: 11/09/2022] Open
Abstract
Excess in mitochondrial reactive oxygen species (ROS) is considered as a major cause of cellular oxidative stress. NADPH, the main intracellular reductant, has a key role in keeping glutathione in its reduced form GSH, which scavenges ROS and thus protects the cell from oxidative damage. Here, we report that SIRT5 desuccinylates and deglutarylates isocitrate dehydrogenase 2 (IDH2) and glucose-6-phosphate dehydrogenase (G6PD), respectively, and thus activates both NADPH-producing enzymes. Moreover, we show that knockdown or knockout of SIRT5 leads to high levels of cellular ROS SIRT5 inactivation leads to the inhibition of IDH2 and G6PD, thereby decreasing NADPH production, lowering GSH, impairing the ability to scavenge ROS, and increasing cellular susceptibility to oxidative stress. Our study uncovers a SIRT5-dependent mechanism that regulates cellular NADPH homeostasis and redox potential by promoting IDH2 desuccinylation and G6PD deglutarylation.
Collapse
Affiliation(s)
- Lisha Zhou
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renqiang Sun
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiufei Chen
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengli Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Xu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shiwen Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue Xiong
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China Department of Biochemistry and Biophysics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kun-Liang Guan
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Pengyuan Yang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongxiu Yu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan Ye
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Fudan University, Shanghai, China Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China Molecular and Cell Biology Lab, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Ravishankar HN, Dutta A, Lewis AG, Mohan A. Advances in therapies and scope for personalized medicine in Alzheimer's disease. Per Med 2016; 13:189-199. [PMID: 29749899 DOI: 10.2217/pme.15.49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer's disease is one of the leading causes of death worldwide and currently does not have any cure. The rate of incidence of Alzheimer's from 2010 is up by 71%, whereas many other diseases have been decreasing in their prevalence. In this review, we have attempted to cover the current landscape of treatment alongside forthcoming advances. We have also covered the present genes identified through genome-wide association studies, which could be used as novel biomarkers and could eventually reduce the cost of treatment through early diagnosis. As this disease is highly polymorphic, applications of personalized medicine have also found its way. All these upcoming developments offer a bright hope in the diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Hosakere N Ravishankar
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| | - Abhik Dutta
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| | - Alisha G Lewis
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| | - Amogh Mohan
- Department of Biotechnology, R.V. College of Engineering, Mysore Road, Bangalore 560059, Karnataka, India
| |
Collapse
|
42
|
McKinney BC, Lin CW, Oh H, Tseng GC, Lewis DA, Sibille E. Hypermethylation of BDNF and SST Genes in the Orbital Frontal Cortex of Older Individuals: A Putative Mechanism for Declining Gene Expression with Age. Neuropsychopharmacology 2015; 40:2604-13. [PMID: 25881116 PMCID: PMC4569950 DOI: 10.1038/npp.2015.107] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/20/2015] [Accepted: 04/08/2015] [Indexed: 12/29/2022]
Abstract
Expression of brain-derived neurotrophic factor (BDNF) and somatostatin (SST) mRNAs in the brain decreases progressively and robustly with age, and lower BDNF and SST expression in the brain has been observed in many brain disorders. BDNF is known to regulate SST expression; however, the mechanisms underlying decreased expression of both genes are not understood. DNA methylation (DNAm) is an attractive candidate mechanism. To investigate the contribution of DNAm to the age-related decline in BDNF and SST expression, the Illumina Infinium HumanMethylation450 Beadchip Array was used to quantify DNAm of BDNF (26 CpG loci) and SST (9 CpG loci) in the orbital frontal cortices of postmortem brains from 22 younger (age <42 years) and 22 older individuals (age >60 years) with known age-dependent BDNF and SST expression differences. Relative to the younger individuals, 10 of the 26 CpG loci in BDNF and 8 of the 9 CpG loci in SST were significantly hypermethylated in the older individuals. DNAm in BDNF exons/promoters I, II, and IV negatively correlated with BDNF expression (r=-0.37, p<0.05; r=-0.40, p<0.05; r=-0.24, p=0.07), and DNAm in SST 5' UTR and first exon/intron negatively correlated with SST expression (r=-0.48, p<0.01; r=-0.63, p<0.001), respectively. An expanded set of BDNF- and GABA-related genes exhibited similar age-related changes in DNAm and correlation with gene expression. These results suggest that DNAm may be a proximal mechanism for decreased expression of BDNF, SST, and other BDNF- and GABA-related genes with brain aging and, by extension, for brain disorders in which their expression is decreased.
Collapse
Affiliation(s)
- Brandon C McKinney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | - Chien-Wei Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hyunjung Oh
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Etienne Sibille
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA,Campbell Family Mental Health Research Institute of CAMH, Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada,Centre for Addiction and Mental Health (CAMH), 250 College Street, Room 134, Toronto, ON M5T 1R8, Canada, Tel: +1 416 535 8501, ext 36571, E-mail:
| |
Collapse
|
43
|
Wong DW, Soga T, Parhar IS. Aging and chronic administration of serotonin-selective reuptake inhibitor citalopram upregulate Sirt4 gene expression in the preoptic area of male mice. Front Genet 2015; 6:281. [PMID: 26442099 PMCID: PMC4584971 DOI: 10.3389/fgene.2015.00281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 08/21/2015] [Indexed: 01/10/2023] Open
Abstract
Sexual dysfunction and cognitive deficits are markers of the aging process. Mammalian sirtuins (SIRT), encoded by sirt 1-7 genes, are known as aging molecules which are sensitive to serotonin (5-hydroxytryptamine, 5-HT). Whether the 5-HT system regulates SIRT in the preoptic area (POA), which could affect reproduction and cognition has not been examined. Therefore, this study was designed to examine the effects of citalopram (CIT, 10 mg/kg for 4 weeks), a potent selective-serotonin reuptake inhibitor and aging on SIRT expression in the POA of male mice using real-time PCR and immunocytochemistry. Age-related increases of sirt1, sirt4, sirt5, and sirt7 mRNA levels were observed in the POA of 52 weeks old mice. Furthermore, 4 weeks of chronic CIT treatment started at 8 weeks of age also increased sirt2 and sirt4 mRNA expression in the POA. Moreover, the number of SIRT4 immuno-reactive neurons increased with aging in the medial septum area (12 weeks = 1.00 ± 0.15 vs. 36 weeks = 1.68 ± 0.14 vs. 52 weeks = 1.54 ± 0.11, p < 0.05). In contrast, the number of sirt4-immunopositive cells did not show a statistically significant change with CIT treatment, suggesting that the increase in sirt4 mRNA levels may occur in cells in which sirt4 is already being expressed. Taken together, these studies suggest that CIT treatment and the process of aging utilize the serotonergic system to up-regulate SIRT4 in the POA as a common pathway to deregulate social cognitive and reproductive functions.
Collapse
Affiliation(s)
- Dutt Way Wong
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia Selangor, Malaysia
| | - Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia Selangor, Malaysia
| | - Ishwar S Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia Selangor, Malaysia
| |
Collapse
|
44
|
Nikolova YS, Iruku SP, Lin CW, Conley ED, Puralewski R, French B, Hariri AR, Sibille E. FRAS1-related extracellular matrix 3 (FREM3) single-nucleotide polymorphism effects on gene expression, amygdala reactivity and perceptual processing speed: An accelerated aging pathway of depression risk. Front Psychol 2015; 6:1377. [PMID: 26441752 PMCID: PMC4584966 DOI: 10.3389/fpsyg.2015.01377] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022] Open
Abstract
The A allele of the FRAS1-related extracellular matrix protein 3 (FREM3) rs7676614 single nucleotide polymorphism (SNP) was linked to major depressive disorder (MDD) in an early genome-wide association study (GWAS), and to symptoms of psychomotor retardation in a follow-up investigation. In line with significant overlap between age- and depression-related molecular pathways, parallel work has shown that FREM3 expression in postmortem human brain decreases with age. Here, we probe the effect of rs7676614 on amygdala reactivity and perceptual processing speed, both of which are altered in depression and aging. Amygdala reactivity was assessed using a face-matching BOLD fMRI paradigm in 365 Caucasian participants in the Duke Neurogenetics Study (DNS) (192 women, mean age 19.7 ± 1.2). Perceptual processing speed was indexed by reaction times in the same task and the Trail Making Test (TMT). The effect of rs7676614 on FREM3 mRNA brain expression levels was probed in a postmortem cohort of 169 Caucasian individuals (44 women, mean age 50.8 ± 14.9). The A allele of rs7676614 was associated with blunted amygdala reactivity to faces, slower reaction times in the face-matching condition (p < 0.04), as well as marginally slower performance on TMT Part B (p = 0.056). In the postmortem cohort, the T allele of rs6537170 (proxy for the rs7676614 A allele), was associated with trend-level reductions in gene expression in Brodmann areas 11 and 47 (p = 0.066), reminiscent of patterns characteristic of older age. The low-expressing allele of another FREM3 SNP (rs1391187) was similarly associated with reduced amygdala reactivity and slower TMT Part B speed, in addition to reduced BA47 activity and extraversion (p < 0.05). Together, these results suggest common genetic variation associated with reduced FREM3 expression may confer risk for a subtype of depression characterized by reduced reactivity to environmental stimuli and slower perceptual processing speed, possibly suggestive of accelerated aging.
Collapse
Affiliation(s)
- Yuliya S. Nikolova
- Campbell Family Mental Health Research Institute of CAMHToronto, ON, Canada
| | - Swetha P. Iruku
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke UniversityDurham, NC, USA
| | - Chien-Wei Lin
- Department of Biostatistics, Graduate School of Public Health, University of PittsburghPittsburgh, PA, USA
| | | | - Rachel Puralewski
- Department of Psychiatry, University of PittsburghPittsburgh, PA, USA
| | - Beverly French
- Department of Psychiatry, University of PittsburghPittsburgh, PA, USA
| | - Ahmad R. Hariri
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke UniversityDurham, NC, USA
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMHToronto, ON, Canada
- Department of Psychiatry, University of PittsburghPittsburgh, PA, USA
- Department of Psychiatry, Department of Pharmacology and Toxicology, University of TorontoToronto, ON, Canada
| |
Collapse
|
45
|
Sood S, Gallagher IJ, Lunnon K, Rullman E, Keohane A, Crossland H, Phillips BE, Cederholm T, Jensen T, van Loon LJC, Lannfelt L, Kraus WE, Atherton PJ, Howard R, Gustafsson T, Hodges A, Timmons JA. A novel multi-tissue RNA diagnostic of healthy ageing relates to cognitive health status. Genome Biol 2015; 16:185. [PMID: 26343147 PMCID: PMC4561473 DOI: 10.1186/s13059-015-0750-x] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/12/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Diagnostics of the human ageing process may help predict future healthcare needs or guide preventative measures for tackling diseases of older age. We take a transcriptomics approach to build the first reproducible multi-tissue RNA expression signature by gene-chip profiling tissue from sedentary normal subjects who reached 65 years of age in good health. RESULTS One hundred and fifty probe-sets form an accurate classifier of young versus older muscle tissue and this healthy ageing RNA classifier performed consistently in independent cohorts of human muscle, skin and brain tissue (n = 594, AUC = 0.83-0.96) and thus represents a biomarker for biological age. Using the Uppsala Longitudinal Study of Adult Men birth-cohort (n = 108) we demonstrate that the RNA classifier is insensitive to confounding lifestyle biomarkers, while greater gene score at age 70 years is independently associated with better renal function at age 82 years and longevity. The gene score is 'up-regulated' in healthy human hippocampus with age, and when applied to blood RNA profiles from two large independent age-matched dementia case-control data sets (n = 717) the healthy controls have significantly greater gene scores than those with cognitive impairment. Alone, or when combined with our previously described prototype Alzheimer disease (AD) RNA 'disease signature', the healthy ageing RNA classifier is diagnostic for AD. CONCLUSIONS We identify a novel and statistically robust multi-tissue RNA signature of human healthy ageing that can act as a diagnostic of future health, using only a peripheral blood sample. This RNA signature has great potential to assist research aimed at finding treatments for and/or management of AD and other ageing-related conditions.
Collapse
Affiliation(s)
- Sanjana Sood
- XRGenomics Ltd, London, UK
- Division of Genetics & Molecular Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Iain J Gallagher
- XRGenomics Ltd, London, UK
- School of Health, Stirling University, Stirling, Scotland, UK
| | - Katie Lunnon
- Department of Old Age Psychiatry, King's College London, London, UK
- Present address: University of Exeter Medical School, Exeter, UK
| | - Eric Rullman
- Division of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Aoife Keohane
- Department of Old Age Psychiatry, King's College London, London, UK
| | - Hannah Crossland
- Division of Genetics & Molecular Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK
- School of Medicine, Derby Royal Hospital, Derbyshire, UK
| | | | - Tommy Cederholm
- Department of Public Health, Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | | | | | - Lars Lannfelt
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - William E Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Robert Howard
- Department of Old Age Psychiatry, King's College London, London, UK
| | - Thomas Gustafsson
- Division of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Angela Hodges
- Department of Old Age Psychiatry, King's College London, London, UK
| | - James A Timmons
- XRGenomics Ltd, London, UK.
- Division of Genetics & Molecular Medicine, King's College London, 8th Floor, Tower Wing, Guy's Hospital, London, SE1 9RT, UK.
| |
Collapse
|
46
|
Aging research-where do we stand and where are we going? Cell 2015; 159:15-19. [PMID: 25259916 DOI: 10.1016/j.cell.2014.08.041] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 08/28/2014] [Accepted: 08/29/2014] [Indexed: 12/26/2022]
Abstract
The 40(th) anniversary of Cell coincides with that of the National Institute on Aging (NIA). Indeed, Cell papers on NIA-funded research helped move the field into a genetic and molecular era. Now is a fair time to ask whether we are far down a trail leading to a deep understanding of aging or whether we are still tiptoeing cautiously at the trailhead.
Collapse
|
47
|
Ghosh S, Zhou Z. SIRTain regulators of premature senescence and accelerated aging. Protein Cell 2015; 6:322-33. [PMID: 25907989 PMCID: PMC4417679 DOI: 10.1007/s13238-015-0149-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/28/2015] [Indexed: 12/11/2022] Open
Abstract
The sirtuin proteins constitute class III histone deacetylases (HDACs). These evolutionarily conserved NAD(+)-dependent enzymes form an important component in a variety of cellular and biological processes with highly divergent as well as convergent roles in maintaining metabolic homeostasis, safeguarding genomic integrity, regulating cancer metabolism and also inflammatory responses. Amongst the seven known mammalian sirtuin proteins, SIRT1 has gained much attention due to its widely acknowledged roles in promoting longevity and ameliorating age-associated pathologies. The contributions of other sirtuins in the field of aging are also gradually emerging. Here, we summarize some of the recent discoveries in sirtuins biology which clearly implicate the functions of sirtuin proteins in the regulation of premature cellular senescence and accelerated aging. The roles of sirtuins in various cellular processes have been extrapolated to draw inter-linkage with anti-aging mechanisms. Also, the latest findings on sirtuins which might have potential effects in the process of aging have been reviewed.
Collapse
Affiliation(s)
- Shrestha Ghosh
- Department of Biochemistry, The University of Hong Kong, Hong Kong, China
| | - Zhongjun Zhou
- Department of Biochemistry, The University of Hong Kong, Hong Kong, China
- Shenzhen Institute of Innovation and Technology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
48
|
Ding Y, Chang LC, Wang X, Guilloux JP, Parrish J, Oh H, French BJ, Lewis DA, Tseng GC, Sibille E. Molecular and Genetic Characterization of Depression: Overlap with other Psychiatric Disorders and Aging. MOLECULAR NEUROPSYCHIATRY 2015. [PMID: 26213687 DOI: 10.1159/000369974] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genome-wide expression and genotyping technologies have uncovered the genetic bases of complex diseases at unprecedented rates; However despite its heavy burden and high prevalence, the molecular characterization of major depressive disorder (MDD) has lagged behind. Transcriptome studies report multiple brain disturbances but are limited by small sample sizes. Genome-wide association studies (GWAS) report weak results but suggest overlapping genetic risk with other neuropsychiatric disorders. We performed systematic molecular characterization of altered brain function in MDD, using meta-analysis of differential expression in eight gene array studies in three corticolimbic brain regions in 101 subjects. The identified "metaA-MDD" genes suggest altered neurotrophic support, brain plasticity and neuronal signaling in MDD. Notably, metaA-MDD genes display low connectivity and hubness in coexpression networks, and uniform genomic distribution, consistent with diffuse polygenic mechanisms. We next integrated these findings with results from over 1800 published GWAS and show that genetic variations nearby metaA-MDD genes predict greater risk for neuropsychiatric disorders and notably for age-related phenotypes, but not for other medical illnesses, including those frequently co-morbid with depression, or body characteristics. Collectively, the intersection of unbiased investigations of gene function (transcriptome) and structure (GWAS) provides novel leads to investigate molecular mechanisms of MDD and suggest common biological pathways between depression, other neuropsychiatric diseases, and brain aging.
Collapse
Affiliation(s)
- Ying Ding
- Joint CMU-Pitt PhD program in Computational Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA ; Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lun-Ching Chang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xingbin Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261, USA ; Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jean-Philippe Guilloux
- Université Paris-Sud EA 3544, Faculté de Pharmacie, Châtenay-Malabry cedex F-92296, France
| | - Jenna Parrish
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15312, USA
| | - Hyunjung Oh
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15312, USA
| | - Beverly J French
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15312, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15312, USA ; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15312, USA
| | - George C Tseng
- Joint CMU-Pitt PhD program in Computational Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA ; Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Etienne Sibille
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15312, USA ; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15312, USA ; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Departments of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, CA
| |
Collapse
|
49
|
Lin CW, Chang LC, Tseng GC, Kirkwood CM, Sibille EL, Sweet RA. VSNL1 Co-Expression Networks in Aging Include Calcium Signaling, Synaptic Plasticity, and Alzheimer's Disease Pathways. Front Psychiatry 2015; 6:30. [PMID: 25806004 PMCID: PMC4353182 DOI: 10.3389/fpsyt.2015.00030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/12/2015] [Indexed: 01/01/2023] Open
Abstract
The visinin-like 1 (VSNL1) gene encodes visinin-like protein 1, a peripheral biomarker for Alzheimer disease (AD). Little is known, however, about normal VSNL1 expression in brain and the biologic networks in which it participates. Frontal cortex gray matter obtained from 209 subjects without neurodegenerative or psychiatric illness, ranging in age from 16 to 91, was processed on Affymetrix GeneChip 1.1 ST and Human SNP Array 6.0. VSNL1 expression was unaffected by age and sex, and not significantly associated with SNPs in cis or trans. VSNL1 was significantly co-expressed with genes in pathways for calcium signaling, AD, long-term potentiation, long-term depression, and trafficking of AMPA receptors. The association with AD was driven, in part, by correlation with amyloid precursor protein (APP) expression. These findings provide an unbiased link between VSNL1 and molecular mechanisms of AD, including pathways implicated in synaptic pathology in AD. Whether APP may drive increased VSNL1 expression, VSNL1 drives increased APP expression, or both are downstream of common pathogenic regulators will need to be evaluated in model systems.
Collapse
Affiliation(s)
- Chien-Wei Lin
- Department of Biostatistics, University of Pittsburgh , Pittsburgh, PA , USA
| | - Lun-Ching Chang
- Department of Biostatistics, University of Pittsburgh , Pittsburgh, PA , USA
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh , Pittsburgh, PA , USA
| | - Caitlin M Kirkwood
- Department of Psychiatry, University of Pittsburgh , Pittsburgh, PA , USA
| | - Etienne L Sibille
- Department of Psychiatry, University of Pittsburgh , Pittsburgh, PA , USA ; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Departments of Psychiatry and Pharmacology & Toxicology, University of Toronto , Toronto, ON , Canada
| | - Robert A Sweet
- Department of Psychiatry, University of Pittsburgh , Pittsburgh, PA , USA ; Department of Neurology, University of Pittsburgh , Pittsburgh, PA , USA ; VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System , Pittsburgh, PA , USA
| |
Collapse
|
50
|
TenNapel MJ, Lynch CF, Burns TL, Wallace R, Smith BJ, Button A, Domann FE. SIRT6 minor allele genotype is associated with >5-year decrease in lifespan in an aged cohort. PLoS One 2014; 9:e115616. [PMID: 25541994 PMCID: PMC4277407 DOI: 10.1371/journal.pone.0115616] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/27/2014] [Indexed: 01/17/2023] Open
Abstract
Aging is a natural process involving complex interplay between environment, metabolism, and genes. Sirtuin genes and their downstream targets have been associated with lifespan in numerous organisms from nematodes to humans. Several target proteins of the sirtuin genes are key sensors and/or effectors of oxidative stress pathways including FOXO3, SOD3, and AKT1. To examine the relationship between single nucleotide polymorphisms (SNP) at candidate genes in these pathways and human lifespan, we performed a molecular epidemiologic study of an elderly cohort (≥65 years old.). Using age at death as a continuous outcome variable and assuming a co-dominant genetic model within the framework of multi-variable linear regression analysis, the genotype-specific adjusted mean age at death was estimated for individual SNP genotypes while controlling for age-related risk factors including smoking, body mass index, alcohol consumption and co-morbidity. Significant associations were detected between human lifespan and SNPs in genes SIRT3, SIRT5, SIRT6, FOXO3 and SOD3. Individuals with either the CC or CT genotype at rs107251 within SIRT6 displayed >5-year mean survival advantages compared to the TT genotype (5.5 and 5.9 years, respectively; q-value = 0.012). Other SNPs revealed genotype-specific mean survival advantages ranging from 0.5 to 1.6 years. Gender also modified the effect of SNPs in SIRT3, SIRT5 and AKT1 on lifespan. Our novel findings highlight the impact of sirtuins and sirtuin-related genotypes on lifespan, the importance of evaluating gender and the advantage of using age as a continuous variable in analyses to report mean age at death.
Collapse
Affiliation(s)
- Mindi J TenNapel
- Department of Epidemiology, College of Public Health, The University of Iowa, 01620 PFPW, 200 Hawkins Drive, Iowa City, Iowa, United States of America
| | - Charles F Lynch
- Department of Epidemiology, College of Public Health, The University of Iowa, 01620 PFPW, 200 Hawkins Drive, Iowa City, Iowa, United States of America
| | - Trudy L Burns
- Department of Epidemiology, College of Public Health, The University of Iowa, 01620 PFPW, 200 Hawkins Drive, Iowa City, Iowa, United States of America
| | - Robert Wallace
- Department of Epidemiology, College of Public Health, The University of Iowa, 01620 PFPW, 200 Hawkins Drive, Iowa City, Iowa, United States of America
| | - Brian J Smith
- Department of Epidemiology, College of Public Health, The University of Iowa, 01620 PFPW, 200 Hawkins Drive, Iowa City, Iowa, United States of America
| | - Anna Button
- Department of Epidemiology, College of Public Health, The University of Iowa, 01620 PFPW, 200 Hawkins Drive, Iowa City, Iowa, United States of America
| | - Frederick E Domann
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|