51
|
Jungert A, Neuhäuser-Berthold M. Determinants of Vitamin B6 Status in Community-Dwelling Older Adults: A Longitudinal Study Over a Period of 18 Years. J Gerontol A Biol Sci Med Sci 2020; 75:374-379. [PMID: 30657862 DOI: 10.1093/gerona/glz010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cross-sectional studies indicate an age-related decline in vitamin B6 status. Because longitudinal studies are lacking, the present study investigates the long-term association between age and vitamin B6 status in older adults by considering potential confounding factors. METHODS The study population consists of 249 women and 111 men aged ≥ 60 years, who had at least three follow-ups between 1996 and 2014 with complete data records on relevant parameters. Vitamin B6 status was assessed by serum pyridoxal 5'-phosphate (PLP) concentrations measured by high-performance liquid chromatography. Linear mixed models were used to analyze the influence of age, sex, body composition, supplements, diet, lifestyle, and serum creatinine on PLP concentrations. RESULTS At baseline, 37% of the subjects showed PLP concentrations < 30 nmol/L and more than half failed to meet the recommended dietary intake. Longitudinal analyses revealed that age, use of supplements and protein intake were positive determinants of PLP concentrations, whereas body fat showed a negative impact. No influence of sex, dietary vitamin B6 intake, lifestyle factors or serum creatinine on PLP concentrations was found. CONCLUSION The present study provides no evidence that in the course of aging PLP concentrations decline between 60 and 90 years. However, age-related changes in body composition, such as an increased ratio of fat mass to fat-free mass may negatively affect vitamin B6 status.
Collapse
Affiliation(s)
- Alexandra Jungert
- Institute of Nutritional Science, Justus Liebig University, Giessen, Germany
| | | |
Collapse
|
52
|
Guo Z, Bao D, Manor B, Zhou J. The Effects of Transcranial Direct Current Stimulation (tDCS) on Balance Control in Older Adults: A Systematic Review and Meta-Analysis. Front Aging Neurosci 2020; 12:275. [PMID: 33024431 PMCID: PMC7516302 DOI: 10.3389/fnagi.2020.00275] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/10/2020] [Indexed: 01/14/2023] Open
Abstract
Background: Recently, considerable research has been conducted to study the effects of transcranial direct current stimulation (tDCS) on balance control in older adults. We completed a comprehensive systematic review and meta-analysis to assess the efficacy of tDCS on balance control in this population. Methods: A search strategy based on the PICOS principle was used to find the literatures in the databases of PubMed, EMBASE, EBSCO, Web of Science. The quality and risk of bias in the studies were independently assessed by two researchers. Results: Ten studies were included in the systematic review. A meta-analysis was completed on six of these ten, with a total of 280 participants. As compared to sham (i.e., control), tDCS induced significant improvement with low heterogeneity in balance control in older adults. Specifically, tDCS induced large effects on the performance of the timed-up-and-go test, the Berg balance scale, and standing postural sway (e.g., sway area) and gait (e.g., walking speed) in dual task conditions (standardized mean differences (SMDs) = -0.99~3.41 95% confidence limits (CL): -1.52~4.50, p < 0.006, I 2 < 52%). Moderate-to-large effects of tDCS were also observed in the standing posture on a static or movable platform (SMDs = 0.37~1.12 95%CL: -0.09~1.62, p < 0.03, I 2 < 62%). Conclusion: Our analysis indicates that tDCS holds promise to promote balance in older adults. These results warrant future studies of larger sample size and rigorous study design and results report, as well as specific research to establish the relationship between the parameter of tDCS and the extent of tDCS-induced improvement in balance control in older adults.
Collapse
Affiliation(s)
- Zhenxiang Guo
- Sports Coaching College, Beijing Sport University, Bejing, China
| | - Dapeng Bao
- China Institute of Sport and Health Science, Beijing Sport University, Bejing, China
| | - Brad Manor
- Hebrew SeniorLife Hinda and Arthur Marcus Institute for Aging Research, Harvard Medical School, Boston, MA, United States
| | - Junhong Zhou
- Hebrew SeniorLife Hinda and Arthur Marcus Institute for Aging Research, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
53
|
Finelli MJ. Redox Post-translational Modifications of Protein Thiols in Brain Aging and Neurodegenerative Conditions-Focus on S-Nitrosation. Front Aging Neurosci 2020; 12:254. [PMID: 33088270 PMCID: PMC7497228 DOI: 10.3389/fnagi.2020.00254] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species and reactive nitrogen species (RONS) are by-products of aerobic metabolism. RONS trigger a signaling cascade that can be transduced through oxidation-reduction (redox)-based post-translational modifications (redox PTMs) of protein thiols. This redox signaling is essential for normal cellular physiology and coordinately regulates the function of redox-sensitive proteins. It plays a particularly important role in the brain, which is a major producer of RONS. Aberrant redox PTMs of protein thiols can impair protein function and are associated with several diseases. This mini review article aims to evaluate the role of redox PTMs of protein thiols, in particular S-nitrosation, in brain aging, and in neurodegenerative diseases. It also discusses the potential of using redox-based therapeutic approaches for neurodegenerative conditions.
Collapse
Affiliation(s)
- Mattéa J Finelli
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
54
|
Rebai A, Reçber T, Nemutlu E, Chbili C, Kurbanoglu S, Kir S, Amor SB, Özkan SA, Saguem S. GC-MS Based Metabolic Profiling of Parkinson's Disease with Glutathione S-transferase M1 and T1 Polymorphism in Tunisian Patients. Comb Chem High Throughput Screen 2020; 23:1041-1048. [PMID: 32342808 DOI: 10.2174/1386207323666200428082815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/13/2020] [Accepted: 04/25/2020] [Indexed: 11/22/2022]
Abstract
AIM AND OBJECTIVE Parkinson's disease (PD) is the second most common neurodegenerative disease. It is a multifactorial disorder (caused by aging, environmental, and genetic factors). Metabolomics can help explore the biomarker profiles for aging. Recent studies showed an association between the glutathione S-transferases (GSTs) polymorphisms and PD risk. The purpose of this study was to evaluate the association of this genetic polymorphism and the metabolomic profile in PD Tunisian patients, in order to identify effective biomarkers in the genetic differentiation. MATERIALS AND METHODS In this study, the metabolomic profile changes related to GSTs polymorphism were searched in 54 Tunisian PD patients treated with L-dopa, using a gas chromatography-mass spectrometry (GC-MS) technique. RESULTS The study results showed that mannose, methyl stearate, and three other unknown metabolites, increased in patients with GSTM1 positive genotype, while glycolic acid, porphine, monomethyl phosphate, fumaric acid, and three other unknown metabolites decreased in patients with GSTM1 positive genotype. Subsequently, the levels of glycolic acid, erythronic acid, lactic acid, citric acid, fructose, stearic acid, 2-amino-2-methyl-1,3-propanediol and three other unknown metabolites increased in patients with GSTM1 positive genotype, while the levels of proline, valine and two unknown metabolites decreased with GSTT1 positive genotype. CONCLUSION All these altered metabolites are related to energy metabolism and it can be concluded that GSTs polymorphism based the shifting in energy metabolism and led to oxidative stress.
Collapse
Affiliation(s)
- Amal Rebai
- Metabolic Biophysics Laboratory, Department of Biophysics, Faculty of Medicine Sousse, Sousse University, Sousse, Tunisia
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Chahra Chbili
- Metabolic Biophysics Laboratory, Department of Biophysics, Faculty of Medicine Sousse, Sousse University, Sousse, Tunisia
| | - Sevinç Kurbanoglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Sedef Kir
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sana B Amor
- Neurology Department of "Sahloul" University Hospital Center (UHC) - Sousse University, Sousse, Tunisia
| | - Sibel A Özkan
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Saad Saguem
- Metabolic Biophysics Laboratory, Department of Biophysics, Faculty of Medicine Sousse, Sousse University, Sousse, Tunisia
| |
Collapse
|
55
|
Islam SJ, Kim JH, Topel M, Liu C, Ko YA, Mujahid MS, Sims M, Mubasher M, Ejaz K, Morgan-Billingslea J, Jones K, Waller EK, Jones D, Uppal K, Dunbar SB, Pemu P, Vaccarino V, Searles CD, Baltrus P, Lewis TT, Quyyumi AA, Taylor H. Cardiovascular Risk and Resilience Among Black Adults: Rationale and Design of the MECA Study. J Am Heart Assoc 2020; 9:e015247. [PMID: 32340530 PMCID: PMC7428584 DOI: 10.1161/jaha.119.015247] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Cardiovascular disease incidence, prevalence, morbidity, and mortality have declined in the past several decades; however, disparities persist among subsets of the population. Notably, blacks have not experienced the same improvements on the whole as whites. Furthermore, frequent reports of relatively poorer health statistics among the black population have led to a broad assumption that black race reliably predicts relatively poorer health outcomes. However, substantial intraethnic and intraracial heterogeneity exists; moreover, individuals with similar risk factors and environmental exposures are often known to experience vastly different cardiovascular health outcomes. Thus, some individuals have good outcomes even in the presence of cardiovascular risk factors, a concept known as resilience. Methods and Results The MECA (Morehouse‐Emory Center for Health Equity) Study was designed to investigate the multilevel exposures that contribute to “resilience” in the face of risk for poor cardiovascular health among blacks in the greater Atlanta, GA, metropolitan area. We used census tract data to determine “at‐risk” and “resilient” neighborhoods with high or low prevalence of cardiovascular morbidity and mortality, based on cardiovascular death, hospitalization, and emergency department visits for blacks. More than 1400 individuals from these census tracts assented to demographic, health, and psychosocial questionnaires administered through telephone surveys. Afterwards, ≈500 individuals were recruited to enroll in a clinical study, where risk biomarkers, such as oxidative stress, and inflammatory markers, endothelial progenitor cells, metabolomic and microRNA profiles, and subclinical vascular dysfunction were measured. In addition, comprehensive behavioral questionnaires were collected and ideal cardiovascular health metrics were assessed using the American Heart Association's Life Simple 7 measure. Last, 150 individuals with low Life Simple 7 were recruited and randomized to a behavioral mobile health (eHealth) plus health coach or eHealth only intervention and followed up for improvement. Conclusions The MECA Study is investigating socioenvironmental and individual behavioral measures that promote resilience to cardiovascular disease in blacks by assessing biological, functional, and molecular mechanisms. REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT03308812.
Collapse
Affiliation(s)
- Shabatun J Islam
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Jeong Hwan Kim
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Matthew Topel
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Chang Liu
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA.,Department of Epidemiology Rollins School of Public Health Emory University Atlanta GA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics Rollins School of Public Health Emory University Atlanta GA
| | - Mahasin S Mujahid
- Division of Epidemiology School of Public Health University of California Berkeley CA
| | - Mario Sims
- Department of Medicine University of Mississippi Medical Center Jackson MS
| | - Mohamed Mubasher
- Department of Community Health and Preventive Medicine Morehouse School of Medicine Atlanta GA
| | - Kiran Ejaz
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Jan Morgan-Billingslea
- Department of Community Health and Preventive Medicine Morehouse School of Medicine Atlanta GA
| | - Kia Jones
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Edmund K Waller
- Department of Hematology and Oncology Winship Cancer Institute Emory University School of Medicine Atlanta GA
| | - Dean Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine Department of Medicine Emory University School of Medicine Atlanta GA
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine Department of Medicine Emory University School of Medicine Atlanta GA
| | - Sandra B Dunbar
- Nell Hodgson Woodruff School of Nursing Emory University Atlanta GA
| | - Priscilla Pemu
- Department of Medicine Morehouse School of Medicine Atlanta GA
| | - Viola Vaccarino
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA.,Department of Epidemiology Rollins School of Public Health Emory University Atlanta GA
| | - Charles D Searles
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Peter Baltrus
- Department of Community Health and Preventive Medicine Morehouse School of Medicine Atlanta GA.,National Center for Primary Care Morehouse School of Medicine Atlanta GA
| | - Tené T Lewis
- Department of Epidemiology Rollins School of Public Health Emory University Atlanta GA
| | - Arshed A Quyyumi
- Division of Cardiology Department of Medicine Emory University School of Medicine Atlanta GA
| | - Herman Taylor
- Department of Medicine Morehouse School of Medicine Atlanta GA
| |
Collapse
|
56
|
Nambo-Venegas R, Valdez-Vargas C, Cisneros B, Palacios-González B, Vela-Amieva M, Ibarra-González I, Cerecedo-Zapata CM, Martínez-Cruz E, Cortés H, Reyes-Grajeda JP, Magaña JJ. Altered Plasma Acylcarnitines and Amino Acids Profile in Spinocerebellar Ataxia Type 7. Biomolecules 2020; 10:biom10030390. [PMID: 32138195 PMCID: PMC7175318 DOI: 10.3390/biom10030390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/19/2022] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7), a neurodegenerative disease characterized by cerebellar ataxia and retinal degeneration, is caused by an abnormal CAG repeat expansion in the ATXN7 gene coding region. The onset and severity of SCA7 are highly variable between patients, thus identification of sensitive biomarkers that accurately diagnose the disease and monitoring its progression are needed. With the aim of identified SCA7-specific metabolites with clinical relevance, we report for the first time, to the best of our knowledge, a metabolomics profiling of circulating acylcarnitines and amino acids in SCA7 patients. We identified 21 metabolites with altered levels in SCA7 patients and determined two different sets of metabolites with diagnostic power. The first signature of metabolites (Valine, Leucine, and Tyrosine) has the ability to discriminate between SCA7 patients and healthy controls, while the second one (Methionine, 3-hydroxytetradecanoyl-carnitine, and 3-hydroxyoctadecanoyl-carnitine) possess the capability to differentiate between early-onset and adult-onset patients, as shown by the multivariate model and ROC analyses. Furthermore, enrichment analyses of metabolic pathways suggest alterations in mitochondrial function, energy metabolism, and fatty acid beta-oxidation in SCA7 patients. In summary, circulating SCA7-specific metabolites identified in this study could serve as effective predictors of SCA7 progression in the clinics, as they are sampled in accessible biofluid and assessed by a relatively simple biochemical assay.
Collapse
Affiliation(s)
- Rafael Nambo-Venegas
- Laboratory of Chronic Diseases Biochemistry, National Genomics Medicine Institute (INMEGEN), Mexico City 14610, Mexico;
| | - Claudia Valdez-Vargas
- Laboratory of Genomic Medicine, Department of Genetics, National Rehabilitation Institute (INR-LGII), Mexico City 14389, Mexico; (C.V.-V.); (H.C.)
- Department of Genetics and Molecular Biology, Center of Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | - Bulmaro Cisneros
- Department of Genetics and Molecular Biology, Center of Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico;
| | | | - Marcela Vela-Amieva
- Laboratory of Inborn errors of metabolism, National Pediatrics Institute (INP), Mexico City 04530, Mexico;
| | | | - César M. Cerecedo-Zapata
- Rehabilitation and Special Education Center of Veracruz (CRISVER-DIF), Xalapa 91097, Veracruz, Mexico; (C.M.C.-Z.)
| | - Emilio Martínez-Cruz
- Rehabilitation and Special Education Center of Veracruz (CRISVER-DIF), Xalapa 91097, Veracruz, Mexico; (C.M.C.-Z.)
| | - Hernán Cortés
- Laboratory of Genomic Medicine, Department of Genetics, National Rehabilitation Institute (INR-LGII), Mexico City 14389, Mexico; (C.V.-V.); (H.C.)
| | - Juan P. Reyes-Grajeda
- Laboratory of Chronic Diseases Biochemistry, National Genomics Medicine Institute (INMEGEN), Mexico City 14610, Mexico;
- Correspondence: (J.P.R.-G.); or (J.J.M.); Tel.: +52-55-5350-1900 (ext. 1192) (J.P.R.-G.); +52-55- 5999-1000 (ext. 14708) (J.J.M.)
| | - Jonathan J. Magaña
- Laboratory of Genomic Medicine, Department of Genetics, National Rehabilitation Institute (INR-LGII), Mexico City 14389, Mexico; (C.V.-V.); (H.C.)
- Correspondence: (J.P.R.-G.); or (J.J.M.); Tel.: +52-55-5350-1900 (ext. 1192) (J.P.R.-G.); +52-55- 5999-1000 (ext. 14708) (J.J.M.)
| |
Collapse
|
57
|
Srivastava S. Emerging Insights into the Metabolic Alterations in Aging Using Metabolomics. Metabolites 2019; 9:E301. [PMID: 31847272 PMCID: PMC6950098 DOI: 10.3390/metabo9120301] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/08/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023] Open
Abstract
Metabolomics is the latest 'omics' technology and systems biology science that allows for comprehensive profiling of small-molecule metabolites in biological systems at a specific time and condition. Metabolites are cellular intermediate products of metabolic reactions, which reflect the ultimate response to genomic, transcriptomic, proteomic, or environmental changes in a biological system. Aging is a complex biological process that is characterized by a gradual and progressive decline in molecular, cellular, tissue, organ, and organismal functions, and it is influenced by a combination of genetic, environmental, diet, and lifestyle factors. The precise biological mechanisms of aging remain unknown. Metabolomics has emerged as a powerful tool to characterize the organism phenotypes, identify altered metabolites, pathways, novel biomarkers in aging and disease, and offers wide clinical applications. Here, I will provide a comprehensive overview of our current knowledge on metabolomics led studies in aging with particular emphasis on studies leading to biomarker discovery. Based on the data obtained from model organisms and humans, it is evident that metabolites associated with amino acids, lipids, carbohydrate, and redox metabolism may serve as biomarkers of aging and/or longevity. Current challenges and key questions that should be addressed in the future to advance our understanding of the biological mechanisms of aging are discussed.
Collapse
Affiliation(s)
- Sarika Srivastava
- Fralin Biomedical Research Institute at Virginia Tech Carilion, 2 Riverside Circle, Roanoke, VA 24016, USA
| |
Collapse
|
58
|
van der Spoel E, van Vliet NA, van Heemst D. Viewpoint on the role of tissue maintenance in ageing: focus on biomarkers of bone, cartilage, muscle, and brain tissue maintenance. Ageing Res Rev 2019; 56:100964. [PMID: 31561015 DOI: 10.1016/j.arr.2019.100964] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/19/2019] [Accepted: 09/19/2019] [Indexed: 12/14/2022]
Abstract
Specific hallmarks are thought to underlie the ageing process and age-related functional decline. In this viewpoint, we put forward the hypothesis that disturbances in the process of tissue maintenance are an important common denominator that may lie in between specific hallmarks of ageing (i.e. damage and responses to damage) and their ultimate (patho)physiological consequences (i.e. functional decline and age-related disease). As a first step towards verifying or falsifying this hypothesis, it will be important to measure biomarkers of tissue maintenance in future studies in different study populations. The main aim of the current paper is to discuss potential biomarkers of tissue maintenance that could be used in such future studies. Among the many tissues that could have been chosen to explore our hypothesis, to keep the paper manageable, we chose to focus on a selected number of tissues, namely bone, cartilage, muscle, and the brain, which are important for mobility and cognition and affected in several common age-related diseases, including osteoporosis, osteoarthritis, sarcopenia, and neurodegenerative diseases. Furthermore, we discuss the advantages and limitations of potential biomarkers for use in (pre)clinical studies. The proposed biomarkers should be validated in future research, for example by measuring these in humans with different rates of ageing.
Collapse
|
59
|
Majidinia M, Karimian A, Alemi F, Yousefi B, Safa A. Targeting miRNAs by polyphenols: Novel therapeutic strategy for aging. Biochem Pharmacol 2019; 173:113688. [PMID: 31682793 DOI: 10.1016/j.bcp.2019.113688] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
Regarding the importance of genetic and epigenetic factors in regulation of aging process, different expression pattern of non-coding RNAs in aging could be investigated. Accordingly, micro RNAs (miRNAs) with a wide range of physiological functions as well as a significant footprint in many diseases have been demonstrated to be down or upregulated during the aging process. Therefore, age-associated microRNAs and their targets have potentially detected the accelerated aging and predicted the risks for age-related diseases. Polyphenols as important antioxidants in human dietary observed in fruits and some beverages have beneficial effects on longevity and aging. Considering miRNAs as an interesting mediator in modulating polyphenols' biological effects, targeting miRNAs which is using polyphenols could be a novel strategy for aging.
Collapse
Affiliation(s)
- Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Ansar Karimian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Student Research Committee, Babol University of medical sciences, Babol, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Amin Safa
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Viet Nam.
| |
Collapse
|
60
|
Wishart DS. Metabolomics for Investigating Physiological and Pathophysiological Processes. Physiol Rev 2019; 99:1819-1875. [PMID: 31434538 DOI: 10.1152/physrev.00035.2018] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Metabolomics uses advanced analytical chemistry techniques to enable the high-throughput characterization of metabolites from cells, organs, tissues, or biofluids. The rapid growth in metabolomics is leading to a renewed interest in metabolism and the role that small molecule metabolites play in many biological processes. As a result, traditional views of metabolites as being simply the "bricks and mortar" of cells or just the fuel for cellular energetics are being upended. Indeed, metabolites appear to have much more varied and far more important roles as signaling molecules, immune modulators, endogenous toxins, and environmental sensors. This review explores how metabolomics is yielding important new insights into a number of important biological and physiological processes. In particular, a major focus is on illustrating how metabolomics and discoveries made through metabolomics are improving our understanding of both normal physiology and the pathophysiology of many diseases. These discoveries are yielding new insights into how metabolites influence organ function, immune function, nutrient sensing, and gut physiology. Collectively, this work is leading to a much more unified and system-wide perspective of biology wherein metabolites, proteins, and genes are understood to interact synergistically to modify the actions and functions of organelles, organs, and organisms.
Collapse
Affiliation(s)
- David S Wishart
- Departments of Biological Sciences and Computing Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
61
|
Trushina E. Alzheimer's disease mechanisms in peripheral cells: Promises and challenges. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:652-660. [PMID: 31720366 PMCID: PMC6838468 DOI: 10.1016/j.trci.2019.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction Development of efficacious therapeutic interventions for Alzheimer's disease (AD) is hampered by the lack of understanding early disease mechanisms, biomarkers, and models that mimic complex pathophysiology of human disease. Methods This article aims to assess to what extent peripheral cells recapitulate molecular mechanisms altered in the brain and could be used as translational models for the development of individualized medicine for AD. Results Multiple studies suggest that AD is a systemic disorder with an active crosstalk between brain and periphery where multiple pathways altered in the brain cells are also affected in plasma, cerebrospinal fluid, and other peripheral cells of AD patients. Discussion Additional studies to validate molecular mechanisms in peripheral cells using advanced system biology techniques and well-characterized cohorts of AD patients together with the development of standardized protocols should be considered to support the application of peripheral cells in AD research.
Collapse
Affiliation(s)
- Eugenia Trushina
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
62
|
Liu X, Zhou QG, Zhu XC, Xie L, Cai BC. Screening for Potential Active Components of Fangji Huangqi Tang on the Treatment of Nephrotic Syndrome by Using Integrated Metabolomics Based on "Correlations Between Chemical and Metabolic Profiles". Front Pharmacol 2019; 10:1261. [PMID: 31695617 PMCID: PMC6817620 DOI: 10.3389/fphar.2019.01261] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022] Open
Abstract
As for traditional Chinese medicine (TCM) prescription, what puzzled researchers most was how to select proper chemical markers to represent the whole pharmacological action system. In this paper, an integrated metabolomic method was presented for a systematic discovery of potential active components in Fangji Huangqi Tang (FHT), a well-known TCM prescription for nephrotic syndrome treatment, based on “correlations between chemical and metabolic profiles.” Firstly, a metabolomics study was carried out to select representative biomarkers of nephrotic syndrome. Then, after drug administration, the dynamic process of serum composition was investigated by the ultra-high performance liquid chromatography coupled with electrospray ionization–quadrupole–time of flight–mass spectrometry (UHPLC-ESI-Q-TOF-MS) technique to detect the prototypes and related metabolites of relative components from FHT. Pearson correlation analysis was finally used to find out the correlations between the endogenous metabolic spectrums and the chemical serum spectrums. As a result, 17 biomarkers for nephrotic syndrome indication were identified, and the main metabolic pathways of their concern included linoleic acid metabolism; cyanoamino acid metabolism; alpha-linolenic acid metabolism; glycine, serine, and threonine metabolism; arachidonic acid metabolism; and glycerophospholipid metabolism. Meanwhile, active components in FHT for nephrotic syndrome treatment were screened out, including (+)-tetrandrine demethylation, fenfangjine G hydrogenation, tetrandrine, N-methylfangchinoline, tetrandrine demethylation, fangchinoline, glycyrrhetic acid, astragaloside II alcohol dehydration, atractylenolide III demethylation + hydrogenation, atractylenolide III demethylation + hydrogenation, and licoricone-N-acetylcysteine conjugation. This study demonstrated a promising way to elucidate the active chemical material basis of TCM prescription.
Collapse
Affiliation(s)
- Xiao Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi-Gang Zhou
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiao-Chai Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Xie
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bao-Chang Cai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
63
|
Silajdžić E, Björkqvist M. A Critical Evaluation of Wet Biomarkers for Huntington's Disease: Current Status and Ways Forward. J Huntingtons Dis 2019; 7:109-135. [PMID: 29614689 PMCID: PMC6004896 DOI: 10.3233/jhd-170273] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
There is an unmet clinical need for objective biomarkers to monitor disease progression and treatment response in Huntington's disease (HD). The aim of this review is, therefore, to provide practical advice for biomarker discovery and to summarise studies on biofluid markers for HD. A PubMed search was performed to review literature with regard to candidate saliva, urine, blood and cerebrospinal fluid biomarkers for HD. Information has been organised into tables to allow a pragmatic approach to the discussion of the evidence and generation of practical recommendations for future studies. Many of the markers published converge on metabolic and inflammatory pathways, although changes in other analytes representing antioxidant and growth factor pathways have also been found. The most promising markers reflect neuronal and glial degeneration, particularly neurofilament light chain. International collaboration to standardise assays and study protocols, as well as to recruit sufficiently large cohorts, will facilitate future biomarker discovery and development.
Collapse
Affiliation(s)
- Edina Silajdžić
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Maria Björkqvist
- Department of Experimental Medical Science, Brain Disease Biomarker Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
64
|
Ha L, Yu M, Yan Z, Rui Z, Zhao B. Effects of Moxibustion and Moxa Smoke on Behavior Changes and Energy Metabolism in APP/PS1 Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:9419567. [PMID: 31485251 PMCID: PMC6710728 DOI: 10.1155/2019/9419567] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/23/2019] [Accepted: 07/17/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the antiaging effects of moxibustion and moxa smoke on APP/PS1 mice and to illustrate the mechanism of moxibustion improving Alzheimer's disease (AD). METHODS 36 male APP/PS1 mice were randomly assigned into three groups (n = 12), including a model control group, a moxibustion group, and a moxa smoke group. In addition, 12 C57BL/6 normal mice served as a normal (negative) control group. Mice in the moxibustion group received moxibustion intervention using Guanyuan (RN4) acupoint. Mice in the moxa smoke group received moxa smoke exposure with the same frequency as the moxibustion group. Behavioral tests were implemented in the 9th week, 3 days after the completion of the intervention. Tricarboxylic acid cycle and fatty acid metabolomics assessments of the mice were determined after behavioral tests. RESULTS In this study, relative to normal mice, we found that AD mice showed altered tricarboxylic and fatty acid metabolism and showed behavioral changes consistent with the onset of AD. However, both the moxibustion and moxa smoke interventions were able to mitigate these effects to some degree in AD mice. CONCLUSIONS The data suggest that tricarboxylic acid cycle and unsaturated fatty acid metabolomics changes may be a target of AD, and the beneficial effects of moxibustion on cognitive behaviors may be mediated by the energy metabolism system.
Collapse
Affiliation(s)
- Lue Ha
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mengyun Yu
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhiyi Yan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhang Rui
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Baixiao Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
65
|
Kofuji S, Hirayama A, Eberhardt AO, Kawaguchi R, Sugiura Y, Sampetrean O, Ikeda Y, Warren M, Sakamoto N, Kitahara S, Yoshino H, Yamashita D, Sumita K, Wolfe K, Lange L, Ikeda S, Shimada H, Minami N, Malhotra A, Morioka S, Ban Y, Asano M, Flanary VL, Ramkissoon A, Chow LML, Kiyokawa J, Mashimo T, Lucey G, Mareninov S, Ozawa T, Onishi N, Okumura K, Terakawa J, Daikoku T, Wise-Draper T, Majd N, Kofuji K, Sasaki M, Mori M, Kanemura Y, Smith EP, Anastasiou D, Wakimoto H, Holland EC, Yong WH, Horbinski C, Nakano I, DeBerardinis RJ, Bachoo RM, Mischel PS, Yasui W, Suematsu M, Saya H, Soga T, Grummt I, Bierhoff H, Sasaki AT. IMP dehydrogenase-2 drives aberrant nucleolar activity and promotes tumorigenesis in glioblastoma. Nat Cell Biol 2019; 21:1003-1014. [PMID: 31371825 PMCID: PMC6686884 DOI: 10.1038/s41556-019-0363-9] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 06/18/2019] [Indexed: 12/17/2022]
Abstract
In many cancers, high proliferation rates correlate with elevation of rRNA and tRNA levels, and nucleolar hypertrophy. However, the underlying mechanisms linking increased nucleolar transcription and tumorigenesis are only minimally understood. Here we show that IMP dehydrogenase-2 (IMPDH2), the rate-limiting enzyme for de novo guanine nucleotide biosynthesis, is overexpressed in the highly lethal brain cancer glioblastoma. This leads to increased rRNA and tRNA synthesis, stabilization of the nucleolar GTP-binding protein nucleostemin, and enlarged, malformed nucleoli. Pharmacological or genetic inactivation of IMPDH2 in glioblastoma reverses these effects and inhibits cell proliferation, whereas untransformed glia cells are unaffected by similar IMPDH2 perturbations. Impairment of IMPDH2 activity triggers nucleolar stress and growth arrest of glioblastoma cells even in the absence of functional p53. Our results reveal that upregulation of IMPDH2 is a prerequisite for the occurance of aberrant nucleolar function and increased anabolic processes in glioblastoma, which constitutes a primary event in gliomagenesis.
Collapse
Affiliation(s)
- Satoshi Kofuji
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Alexander Otto Eberhardt
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Risa Kawaguchi
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Oltea Sampetrean
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiki Ikeda
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mikako Warren
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Naoya Sakamoto
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuji Kitahara
- Department of Anatomy and Developmental Biology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Hirofumi Yoshino
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kazutaka Sumita
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kara Wolfe
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lisa Lange
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Satsuki Ikeda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Hiroko Shimada
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Noriaki Minami
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Akshiv Malhotra
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shin Morioka
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Ban
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Maya Asano
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Victoria L Flanary
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Annmarie Ramkissoon
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lionel M L Chow
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Juri Kiyokawa
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomoyuki Mashimo
- Department of Internal Medicine; Harold C. Simmons Comprehensive Cancer Center; Annette G. Strauss Center for Neuro-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Greg Lucey
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sergey Mareninov
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tatsuya Ozawa
- Division of Human Biology, Solid Tumor and Translational Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nobuyuki Onishi
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Okumura
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jumpei Terakawa
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, Kanazawa, Japan
| | - Takiko Daikoku
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, Kanazawa, Japan
| | - Trisha Wise-Draper
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nazanin Majd
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kaori Kofuji
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mika Sasaki
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Masaru Mori
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Eric P Smith
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eric C Holland
- Division of Human Biology, Solid Tumor and Translational Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - William H Yong
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Craig Horbinski
- Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA
- Departments of Pathology and Neurosurgery, Northwestern University, Chicago, IL, USA
| | - Ichiro Nakano
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute; Children's Medical Center Research Institute; Department of Pediatrics and Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Robert M Bachoo
- Department of Internal Medicine; Harold C. Simmons Comprehensive Cancer Center; Annette G. Strauss Center for Neuro-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research; Department of Pathology; Moores Cancer Center, University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Wataru Yasui
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- AMED-CREST, AMED, Tokyo, Japan
| | - Ingrid Grummt
- Division of Molecular Biology of the Cell II, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Holger Bierhoff
- Institute of Biochemistry and Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena, Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Atsuo T Sasaki
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan.
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Department of Neurosurgery, Brain Tumor Center at UC Gardner Neuroscience Institute, Cincinnati, OH, USA.
| |
Collapse
|
66
|
Li Y, Liu L, Tian Y, Zhang J. Rapamycin improves sevoflurane‑induced cognitive dysfunction in aged rats by mediating autophagy through the TLR4/MyD88/NF‑κB signaling pathway. Mol Med Rep 2019; 20:3085-3094. [PMID: 31432123 PMCID: PMC6755174 DOI: 10.3892/mmr.2019.10541] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/20/2019] [Indexed: 12/25/2022] Open
Abstract
The present study was aimed to observe the protective effect of rapamycin on cognitive dysfunction induced by sevoflurane in aged rats and its effect on autophagy-related proteins, and to investigate the regulatory mechanism of the Toll-like receptor 4/myeloid differentiation primary response 88/nuclear factor-κB (TLR4/MyD88/NF-κB) signaling pathway. Fifty Sprague-Dawley rats were randomly assigned to a control group, a sevoflurane group, a rapamycin pretreatment group, a TLR4 inhibitor group and a 3MA autophagy inhibitor group. A water maze test was used to evaluate the cognition and memory of rats. Hematoxylin and eosin (H&E) staining was performed to observe pathological changes of brain tissue. A TUNEL assay was used to detect the apoptosis of brain tissue. ELISA was used to assess changes in brain injury markers and inflammatory factors. A western blot assay or quantitative reverse transcription PCR (RT-qPCR) were performed to determine the expression of autophagy-related proteins and the TLR4/MyD88/NF-κB signaling pathway in brain tissue. The results revealed that rapamycin could improve cognitive dysfunction of aged rats induced by sevoflurane. Rapamycin was identified to play a therapeutic role, including mitigating brain tissue damage, inhibiting apoptosis, and activating autophagy in a sevoflurane-treated aged rat model. This function of rapamycin was demonstrated to depend on the TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Lidan Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yue Tian
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
67
|
Laíns I, Chung W, Kelly RS, Gil J, Marques M, Barreto P, Murta JN, Kim IK, Vavvas DG, Miller JB, Silva R, Lasky-Su J, Liang L, Miller JW, Husain D. Human Plasma Metabolomics in Age-Related Macular Degeneration: Meta-Analysis of Two Cohorts. Metabolites 2019; 9:E127. [PMID: 31269701 PMCID: PMC6680405 DOI: 10.3390/metabo9070127] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 01/01/2023] Open
Abstract
The pathogenesis of age-related macular degeneration (AMD), a leading cause of blindness worldwide, remains only partially understood. This has led to the current lack of accessible and reliable biofluid biomarkers for diagnosis and prognosis, and absence of treatments for dry AMD. This study aimed to assess the plasma metabolomic profiles of AMD and its severity stages with the ultimate goal of contributing to addressing these needs. We recruited two cohorts: Boston, United States (n = 196) and Coimbra, Portugal (n = 295). Fasting blood samples were analyzed using ultra-high performance liquid chromatography mass spectrometry. For each cohort, we compared plasma metabolites of AMD patients versus controls (logistic regression), and across disease stages (permutation-based cumulative logistic regression considering both eyes). Meta-analyses were then used to combine results from the two cohorts. Our results revealed that 28 metabolites differed significantly between AMD patients versus controls (false discovery rate (FDR) q-value: 4.1 × 10-2-1.8 × 10-5), and 67 across disease stages (FDR q-value: 4.5 × 10-2-1.7 × 10-4). Pathway analysis showed significant enrichment of glycerophospholipid, purine, taurine and hypotaurine, and nitrogen metabolism (p-value ≤ 0.04). In conclusion, our findings support that AMD patients present distinct plasma metabolomic profiles, which vary with disease severity. This work contributes to the understanding of AMD pathophysiology, and can be the basis of future biomarkers and precision medicine for this blinding condition.
Collapse
Affiliation(s)
- Inês Laíns
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, 3000 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, 3000 Coimbra, Portugal
| | - Wonil Chung
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Rachel S Kelly
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - João Gil
- Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal
| | - Marco Marques
- Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal
| | - Patrícia Barreto
- Association for Innovation and Biomedical Research on Light and Image, 3000 Coimbra, Portugal
| | - Joaquim N Murta
- Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, 3000 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, 3000 Coimbra, Portugal
| | - Ivana K Kim
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G Vavvas
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - John B Miller
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Rufino Silva
- Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, 3000 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, 3000 Coimbra, Portugal
| | - Jessica Lasky-Su
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Liming Liang
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Joan W Miller
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Deeba Husain
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
68
|
Yang ZH, Shi CH, Zhou LN, Li YS, Yang J, Liu YT, Mao CY, Luo HY, Xu GW, Xu YM. Metabolic Profiling Reveals Biochemical Pathways and Potential Biomarkers of Spinocerebellar Ataxia 3. Front Mol Neurosci 2019; 12:159. [PMID: 31316347 PMCID: PMC6611058 DOI: 10.3389/fnmol.2019.00159] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022] Open
Abstract
Spinocerebellar ataxia 3, also known as Machado-Joseph disease (SCA3/MJD), is a rare autosomal-dominant neurodegenerative disease caused by an abnormal expansion of CAG repeats in the ATXN3 gene. In the present study, we performed a global metabolomic analysis to identify pathogenic biochemical pathways and novel biomarkers implicated in SCA3 patients. Metabolic profiling of serum samples from 13 preclinical SCA3 patients, 13 symptomatic SCA3 patients, and 15 healthy controls were mapped using ultra-high-performance liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry techniques. The symptomatic SCA3 patients showed a metabolic profile significantly distinct from those of the preclinical SCA3 patients and healthy controls. The principal differential metabolites were involved in the amino acid (AA) metabolism and fatty acid metabolism pathways. In addition, four candidate serum biomarkers, FFA 16:1 (palmitoleic acid), FFA 18:3 (linolenic acid), L-Proline and L-Tryptophan, were selected to discriminate between symptomatic SCA3 patients and healthy controls by receiver operator curve analysis with an area under the curve of 0.979. Our study demonstrates that symptomatic SCA3 patients present distinct metabolic profiles with perturbed AA metabolism and fatty acid metabolism, and FFA 16:1, FFA 18:3, L-Proline and L-Tryptophan are identified as potential disease biomarkers.
Collapse
Affiliation(s)
- Zhi-Hua Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Chang-He Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Li-Na Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yu-Sheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yu-Tao Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Cheng-Yuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Hai-Yang Luo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Guo-Wang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
69
|
Durani LW, Hamezah HS, Ibrahim NF, Yanagisawa D, Nasaruddin ML, Mori M, Azizan KA, Damanhuri HA, Makpol S, Wan Ngah WZ, Tooyama I. Tocotrienol-Rich Fraction of Palm Oil Improves Behavioral Impairments and Regulates Metabolic Pathways in AβPP/PS1 Mice. J Alzheimers Dis 2019; 64:249-267. [PMID: 29889072 PMCID: PMC6004929 DOI: 10.3233/jad-170880] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have recently shown that the tocotrienol-rich fraction (TRF) of palm oil, a mixture of vitamin E analogs, improves amyloid pathology in vitro and in vivo. However, precise mechanisms remain unknown. In this study, we examined the effects of long-term (10 months) TRF treatment on behavioral impairments and brain metabolites in (15 months old) AβPP/PS1 double transgenic (Tg) Alzheimer’s disease (AD) mice. The open field test, Morris water maze, and novel object recognition tasks revealed improved exploratory activity, spatial learning, and recognition memory, respectively, in TRF-treated Tg mice. Brain metabolite profiling of wild-type and Tg mice treated with and without TRF was performed using ultrahigh performance liquid chromatography (UHPLC) coupled to high-resolution accurate mass (HRAM)-orbitrap tandem mass spectrometry (MS/MS). Metabolic pathway analysis found perturbed metabolic pathways that linked to AD. TRF treatment partly ameliorated metabolic perturbations in Tg mouse hippocampus. The mechanism of this pre-emptive activity may occur via modulation of metabolic pathways dependent on Aβ interaction or independent of Aβ interaction.
Collapse
Affiliation(s)
- Lina Wati Durani
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Hamizah Shahirah Hamezah
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Nor Faeizah Ibrahim
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Muhammad Luqman Nasaruddin
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Masaki Mori
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| | - Kamalrul Azlan Azizan
- Metabolomics Research Laboratory, Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia (UKM), Bangi, Selangor, Malaysia
| | - Hanafi Ahmad Damanhuri
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Wan Zurinah Wan Ngah
- Department of Biochemistry, Faculty of Medicine, UKMMC, Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Japan
| |
Collapse
|
70
|
Majuta SN, Li C, Jayasundara K, Kiani Karanji A, Attanayake K, Ranganathan N, Li P, Valentine SJ. Rapid Solution-Phase Hydrogen/Deuterium Exchange for Metabolite Compound Identification. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:1102-1114. [PMID: 30980382 DOI: 10.1007/s13361-019-02163-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 05/25/2023]
Abstract
Rapid, solution-phase hydrogen/deuterium exchange (HDX) coupled with mass spectrometry (MS) is demonstrated as a means for distinguishing small-molecule metabolites. HDX is achieved using capillary vibrating sharp-edge spray ionization (cVSSI) to allow sufficient time for reagent mixing and exchange in micrometer-sized droplets. Different compounds are observed to incorporate deuterium with varying efficiencies resulting in unique isotopic patterns as revealed in the MS spectra. Using linear regression techniques, parameters representing contribution to exchange by different hydrogen types can be computed. In this proof-of-concept study, the exchange parameters are shown to be useful in the retrodiction of the amount of deuterium incorporated within different compounds. On average, the exchange parameters retrodict the exchange level with ~ 2.2-fold greater accuracy than treating all exchangeable hydrogens equally. The parameters can be used to produce hypothetical isotopic distributions that agree (± 16% RMSD) with experimental measurements. These initial studies are discussed in light of their potential value for identifying challenging metabolite species.
Collapse
Affiliation(s)
- Sandra N Majuta
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Chong Li
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Kinkini Jayasundara
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Ahmad Kiani Karanji
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Kushani Attanayake
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Nandhini Ranganathan
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Peng Li
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Stephen J Valentine
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
71
|
Servià L, Jové M, Sol J, Pamplona R, Badia M, Montserrat N, Portero-Otin M, Trujillano J. A prospective pilot study using metabolomics discloses specific fatty acid, catecholamine and tryptophan metabolic pathways as possible predictors for a negative outcome after severe trauma. Scand J Trauma Resusc Emerg Med 2019; 27:56. [PMID: 31118076 PMCID: PMC6530007 DOI: 10.1186/s13049-019-0631-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022] Open
Abstract
Background We wanted to define metabolomic patterns in plasma to predict a negative outcome in severe trauma patients. Methods A prospective pilot study was designed to evaluate plasma metabolomic patterns, established by liquid chromatography coupled to mass spectrometry, in patients allocated to an intensive care unit (in the University Hospital Arnau de Vilanova, Lleida, Spain) in the first hours after a severe trauma (n = 48). Univariate and multivariate statistics were employed to establish potential predictors of mortality. Results Plasma of patients non surviving to trauma (n = 5) exhibited a discriminating metabolomic pattern, involving basically metabolites belonging to fatty acid and catecholamine synthesis as well as tryptophan degradation pathways. Thus, concentration of several metabolites exhibited an area under the receiver operating curve (ROC) higher than 0.84, including 3-indolelactic acid, hydroxyisovaleric acid, phenylethanolamine, cortisol, epinephrine and myristic acid. Multivariate binary regression logistic revealed that patients with higher myristic acid concentrations had a non-survival odds ratio of 2.1 (CI 95% 1.1–3.9). Conclusions Specific fatty acids, catecholamine synthesis and tryptophan degradation pathways could be implicated in a negative outcome after trauma. The metabolomic study of severe trauma patients could be helpful for biomarker proposal. Electronic supplementary material The online version of this article (10.1186/s13049-019-0631-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Luis Servià
- Critical Care Unit, University Hospital Arnau de Vilanova, 25198, Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Joaquim Sol
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain
| | - Mariona Badia
- Critical Care Unit, University Hospital Arnau de Vilanova, 25198, Lleida, Spain
| | - Neus Montserrat
- Critical Care Unit, University Hospital Arnau de Vilanova, 25198, Lleida, Spain
| | - Manuel Portero-Otin
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (IRBLleida), 25198, Lleida, Spain.
| | - Javier Trujillano
- Critical Care Unit, University Hospital Arnau de Vilanova, 25198, Lleida, Spain.
| |
Collapse
|
72
|
Lima Giacobbo B, Doorduin J, Klein HC, Dierckx RAJO, Bromberg E, de Vries EFJ. Brain-Derived Neurotrophic Factor in Brain Disorders: Focus on Neuroinflammation. Mol Neurobiol 2019; 56:3295-3312. [PMID: 30117106 PMCID: PMC6476855 DOI: 10.1007/s12035-018-1283-6] [Citation(s) in RCA: 483] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/24/2018] [Indexed: 12/26/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most studied neurotrophins in the healthy and diseased brain. As a result, there is a large body of evidence that associates BDNF with neuronal maintenance, neuronal survival, plasticity, and neurotransmitter regulation. Patients with psychiatric and neurodegenerative disorders often have reduced BDNF concentrations in their blood and brain. A current hypothesis suggests that these abnormal BDNF levels might be due to the chronic inflammatory state of the brain in certain disorders, as neuroinflammation is known to affect several BDNF-related signaling pathways. Activation of glia cells can induce an increase in the levels of pro- and antiinflammatory cytokines and reactive oxygen species, which can lead to the modulation of neuronal function and neurotoxicity observed in several brain pathologies. Understanding how neuroinflammation is involved in disorders of the brain, especially in the disease onset and progression, can be crucial for the development of new strategies of treatment. Despite the increasing evidence for the involvement of BDNF and neuroinflammation in brain disorders, there is scarce evidence that addresses the interaction between the neurotrophin and neuroinflammation in psychiatric and neurodegenerative diseases. This review focuses on the effect of acute and chronic inflammation on BDNF levels in the most common psychiatric and neurodegenerative disorders and aims to shed some light on the possible biological mechanisms that may influence this effect. In addition, this review will address the effect of behavior and pharmacological interventions on BDNF levels in these disorders.
Collapse
Affiliation(s)
- Bruno Lima Giacobbo
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, Porto Alegre, 90619-900, Brazil
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 31.001, 9713 GZ, Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 31.001, 9713 GZ, Groningen, The Netherlands
| | - Hans C Klein
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 31.001, 9713 GZ, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 31.001, 9713 GZ, Groningen, The Netherlands
| | - Elke Bromberg
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Ipiranga Av. 6681, Porto Alegre, 90619-900, Brazil
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 31.001, 9713 GZ, Groningen, The Netherlands.
| |
Collapse
|
73
|
Jiang Y, Zhu Z, Shi J, An Y, Zhang K, Wang Y, Li S, Jin L, Ye W, Cui M, Chen X. Metabolomics in the Development and Progression of Dementia: A Systematic Review. Front Neurosci 2019; 13:343. [PMID: 31031585 PMCID: PMC6474157 DOI: 10.3389/fnins.2019.00343] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
Dementia has become a major global public health challenge with a heavy economic burden. It is urgently necessary to understand dementia pathogenesis and to identify biomarkers predicting risk of dementia in the preclinical stage for prevention, monitoring, and treatment. Metabolomics provides a novel approach for the identification of biomarkers of dementia. This systematic review aimed to examine and summarize recent retrospective cohort human studies assessing circulating metabolite markers, detected using high-throughput metabolomics, in the context of disease progression to dementia, including incident mild cognitive impairment, all-cause dementia, and cognitive decline. We systematically searched the PubMed, Embase, and Cochrane databases for retrospective cohort human studies assessing associations between blood (plasma or serum) metabolomics profile and cognitive decline and risk of dementia from inception through October 15, 2018. We identified 16 studies reporting circulating metabolites and risk of dementia, and six regarding cognitive performance change. Concentrations of several blood metabolites, including lipids (higher phosphatidylcholines, sphingomyelins, and lysophophatidylcholine, and lower docosahexaenoic acid and high-density lipoprotein subfractions), amino acids (lower branched-chain amino acids, creatinine, and taurine, and higher glutamate, glutamine, and anthranilic acid), and steroids were associated with cognitive decline and the incidence or progression of dementia. Circulating metabolites appear to be associated with the risk of dementia. Metabolomics could be a promising tool in dementia biomarker discovery. However, standardization and consensus guidelines for study design and analytical techniques require future development.
Collapse
Affiliation(s)
- Yanfeng Jiang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Zhen Zhu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Key Laboratory of Public Health Safety of Ministry of Education, Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Jie Shi
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanpeng An
- State Key Laboratory of Genetic Engineering, Metabonomics and Systems Biology Laboratory, School of Life Sciences, Fudan University, Shanghai, China
| | - Kexun Zhang
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Key Laboratory of Public Health Safety of Ministry of Education, Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Yingzhe Wang
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuyuan Li
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Human Phenome Institute, Fudan University, Shanghai, China
| | - Weimin Ye
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mei Cui
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Fudan University Taizhou Institute of Health Sciences, Taizhou, China.,Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
74
|
Abstract
Glucose is the long-established, obligatory fuel for brain that fulfills many critical functions, including ATP production, oxidative stress management, and synthesis of neurotransmitters, neuromodulators, and structural components. Neuronal glucose oxidation exceeds that in astrocytes, but both rates increase in direct proportion to excitatory neurotransmission; signaling and metabolism are closely coupled at the local level. Exact details of neuron-astrocyte glutamate-glutamine cycling remain to be established, and the specific roles of glucose and lactate in the cellular energetics of these processes are debated. Glycolysis is preferentially upregulated during brain activation even though oxygen availability is sufficient (aerobic glycolysis). Three major pathways, glycolysis, pentose phosphate shunt, and glycogen turnover, contribute to utilization of glucose in excess of oxygen, and adrenergic regulation of aerobic glycolysis draws attention to astrocytic metabolism, particularly glycogen turnover, which has a high impact on the oxygen-carbohydrate mismatch. Aerobic glycolysis is proposed to be predominant in young children and specific brain regions, but re-evaluation of data is necessary. Shuttling of glucose- and glycogen-derived lactate from astrocytes to neurons during activation, neurotransmission, and memory consolidation are controversial topics for which alternative mechanisms are proposed. Nutritional therapy and vagus nerve stimulation are translational bridges from metabolism to clinical treatment of diverse brain disorders.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences , Little Rock, Arkansas ; and Department of Cell Biology and Physiology, University of New Mexico , Albuquerque, New Mexico
| |
Collapse
|
75
|
Maniati MS, Maniati M, Yousefi T, Ahmadi‐Ahangar A, Tehrani SS. New insights into the role of microRNAs and long noncoding RNAs in most common neurodegenerative diseases. J Cell Biochem 2019; 120:8908-8918. [DOI: 10.1002/jcb.28361] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/29/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Mohammad Saeed Maniati
- Student Research Committee, Babol University of Medical Sciences Babol Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences Babol Iran
| | - Mahmood Maniati
- Department of English Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Tooba Yousefi
- Student Research Committee, Babol University of Medical Sciences Babol Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences Babol Iran
| | - Alijan Ahmadi‐Ahangar
- Mobility Impairment Research Center, Babol University of Medical Sciences Babol Iran
| | - Sadra Samavarchi Tehrani
- Student Research Committee, Babol University of Medical Sciences Babol Iran
- Mobility Impairment Research Center, Babol University of Medical Sciences Babol Iran
| |
Collapse
|
76
|
Liao WT, Liu J, Zhou SM, Xu G, Gao YQ, Liu WY. UHPLC-QTOFMS-Based Metabolomic Analysis of the Hippocampus in Hypoxia Preconditioned Mouse. Front Physiol 2019; 9:1950. [PMID: 30687133 PMCID: PMC6335317 DOI: 10.3389/fphys.2018.01950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/22/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Hypoxia appears in a number of extreme environments, including high altitudes, the deep sea, and during aviation, and occurs in cancer, cardiovascular disease, respiratory failures and neurological disorders. Though it is well recognized that hypoxic preconditioning (HPC) exerts endogenous neuroprotective effect against severe hypoxia, the mediators and underlying molecular mechanism for the protective effect are still not fully understood. This study established a hippocampus metabolomics approach to explore the alterations associated with HPC. Methods: In this study, an animal model of HPC was established by exposing the adult BALB/c mice to acute repetitive hypoxia four times. Ultra-high liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-QTOFMS) in combination with univariate and multivariate statistical analyses was employed to deciphering metabolic changes associated with HPC in hippocampus tissue. MetaboAnalyst 3.0 was used to construct HPC related metabolic pathways. Results: The significant metabolic differences in hippocampus between the HPC groups and control were observed, indicating that HPC mouse model was successfully established and HPC could caused significant metabolic changes. Several key metabolic pathways were found to be acutely perturbed, including phenylalanine, tyrosine and tryptophan biosynthesis, taurine and hypotaurine metabolism, phenylalanine metabolism, glutathione metabolism, alanine, aspartate and glutamate metabolism, tyrosine metabolism, tryptophan metabolism, purine metabolism, citrate cycle, and glycerophospholipid metabolism. Conclusion: The results of the present study provided novel insights into the mechanisms involved in the acclimatization of organisms to hypoxia, and demonstrated the neuroprotective mechanism of HPC.
Collapse
Affiliation(s)
- Wen-Ting Liao
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China
| | - Jie Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China.,The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Si-Min Zhou
- The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China.,Department of High Altitude Military Hygiene, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China.,The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China.,The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Wen-Yuan Liu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
77
|
Pickett SB, Thomas ED, Sebe JY, Linbo T, Esterberg R, Hailey DW, Raible DW. Cumulative mitochondrial activity correlates with ototoxin susceptibility in zebrafish mechanosensory hair cells. eLife 2018; 7:38062. [PMID: 30596476 PMCID: PMC6345563 DOI: 10.7554/elife.38062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 12/31/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a prominent role in mechanosensory hair cell damage and death. Although hair cells are thought to be energetically demanding cells, how mitochondria respond to these demands and how this might relate to cell death is largely unexplored. Using genetically encoded indicators, we found that mitochondrial calcium flux and oxidation are regulated by mechanotransduction and demonstrate that hair cell activity has both acute and long-term consequences on mitochondrial function. We tested whether variation in mitochondrial activity reflected differences in the vulnerability of hair cells to the toxic drug neomycin. We observed that susceptibility did not correspond to the acute level of mitochondrial activity but rather to the cumulative history of that activity.
Collapse
Affiliation(s)
- Sarah B Pickett
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Eric D Thomas
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Joy Y Sebe
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Tor Linbo
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Robert Esterberg
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - Dale W Hailey
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| |
Collapse
|
78
|
Jové M, Pradas I, Dominguez-Gonzalez M, Ferrer I, Pamplona R. Lipids and lipoxidation in human brain aging. Mitochondrial ATP-synthase as a key lipoxidation target. Redox Biol 2018; 23:101082. [PMID: 30635167 PMCID: PMC6859548 DOI: 10.1016/j.redox.2018.101082] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/04/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022] Open
Abstract
The human brain is a target of the aging process like other cell systems of the human body. Specific regions of the human brain exhibit differential vulnerabilities to the aging process. Yet the underlying mechanisms that sustain the preservation or deterioration of neurons and cerebral functions are unknown. In this review, we focus attention on the role of lipids and the importance of the cross-regionally different vulnerabilities in human brain aging. In particular, we first consider a brief approach to the lipidomics of human brain, the relationship between lipids and lipoxidative damage, the role of lipids in human brain aging, and the specific targets of lipoxidative damage in human brain and during aging. It is proposed that the restricted set of modified proteins and the functional categories involved may be considered putative collaborative factors contributing to neuronal aging, and that mitochondrial ATP synthase is a key lipoxidative target in human brain aging.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain
| | - Irene Pradas
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain
| | - Mayelin Dominguez-Gonzalez
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), ISCIII, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain.
| |
Collapse
|
79
|
Laíns I, Gantner M, Murinello S, Lasky-Su JA, Miller JW, Friedlander M, Husain D. Metabolomics in the study of retinal health and disease. Prog Retin Eye Res 2018; 69:57-79. [PMID: 30423446 DOI: 10.1016/j.preteyeres.2018.11.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/06/2018] [Accepted: 11/07/2018] [Indexed: 02/06/2023]
Abstract
Metabolomics is the qualitative and quantitative assessment of the metabolites (small molecules < 1.5 kDa) in body fluids. The metabolites are the downstream of the genetic transcription and translation processes and also downstream of the interactions with environmental exposures; thus, they are thought to closely relate to the phenotype, especially for multifactorial diseases. In the last decade, metabolomics has been increasingly used to identify biomarkers in disease, and it is currently recognized as a very powerful tool with great potential for clinical translation. The metabolome and the associated pathways also help improve our understanding of the pathophysiology and mechanisms of disease. While there has been increasing interest and research in metabolomics of the eye, the application of metabolomics to retinal diseases has been limited, even though these are leading causes of blindness. In this manuscript, we perform a comprehensive summary of the tools and knowledge required to perform a metabolomics study, and we highlight essential statistical methods for rigorous study design and data analysis. We review available protocols, summarize the best approaches, and address the current unmet need for information on collection and processing of tissues and biofluids that can be used for metabolomics of retinal diseases. Additionally, we critically analyze recent work in this field, both in animal models and in human clinical disease, including diabetic retinopathy and age-related macular degeneration. Finally, we identify opportunities for future research applying metabolomics to improve our current assessment and understanding of mechanisms of vitreoretinal diseases, and to hence improve patient assessment and care.
Collapse
Affiliation(s)
- Inês Laíns
- Retina Service, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, United States; Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal.
| | - Mari Gantner
- Lowy Medical Research Institute, La Jolla, CA, 92037, United States; Scripps Research Institute, La Jolla, CA, 92037, United States.
| | - Salome Murinello
- Lowy Medical Research Institute, La Jolla, CA, 92037, United States; Scripps Research Institute, La Jolla, CA, 92037, United States.
| | - Jessica A Lasky-Su
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, United States.
| | - Joan W Miller
- Retina Service, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, United States.
| | - Martin Friedlander
- Lowy Medical Research Institute, La Jolla, CA, 92037, United States; Scripps Research Institute, La Jolla, CA, 92037, United States.
| | - Deeba Husain
- Retina Service, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, United States.
| |
Collapse
|
80
|
Sapkota S, Huan T, Tran T, Zheng J, Camicioli R, Li L, Dixon RA. Alzheimer's Biomarkers From Multiple Modalities Selectively Discriminate Clinical Status: Relative Importance of Salivary Metabolomics Panels, Genetic, Lifestyle, Cognitive, Functional Health and Demographic Risk Markers. Front Aging Neurosci 2018; 10:296. [PMID: 30333744 PMCID: PMC6175993 DOI: 10.3389/fnagi.2018.00296] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Among the neurodegenerative diseases of aging, sporadic Alzheimer’s disease (AD) is the most prevalent and perhaps the most feared. With virtually no success at finding pharmaceutical therapeutics for altering progressive AD after diagnosis, research attention is increasingly directed at discovering biological and other markers that detect AD risk in the long asymptomatic phase. Both early detection and precision preclinical intervention require systematic investigation of multiple modalities and combinations of AD-related biomarkers and risk factors. We extend recent unbiased metabolomics research that produced a set of metabolite biomarker panels tailored to the discrimination of cognitively normal (CN), cognitively impaired and AD patients. Specifically, we compare the prediction importance of these panels with five other sets of modifiable and non-modifiable AD risk factors (genetic, lifestyle, cognitive, functional health and bio-demographic) in three clinical groups. Method: The three groups were: CN (n = 35), mild cognitive impairment (MCI; n = 25), and AD (n = 22). In a series of three pairwise comparisons, we used machine learning technology random forest analysis (RFA) to test relative predictive importance of up to 19 risk biomarkers from the six AD risk domains. Results: The three RFA multimodal prediction analyses produced significant discriminating risk factors. First, discriminating AD from CN was the AD metabolite panel and two cognitive markers. Second, discriminating AD from MCI was the AD/MCI metabolite panel and two cognitive markers. Third, discriminating MCI from CN was the MCI metabolite panel and seven markers from four other risk modalities: genetic, lifestyle, cognition and functional health. Conclusions: Salivary metabolomics biomarker panels, supplemented by other risk markers, were robust predictors of: (1) clinical differences in impairment and dementia and even; (2) subtle differences between CN and MCI. For the latter, the metabolite panel was supplemented by biomarkers that were both modifiable (e.g., functional) and non-modifiable (e.g., genetic). Comparing, integrating and identifying important multi-modal predictors may lead to novel combinations of complex risk profiles potentially indicative of neuropathological changes in asymptomatic or preclinical AD.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Tao Huan
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Tran Tran
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jiamin Zheng
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.,Department of Medicine (Neurology), University of Alberta, Edmonton, AB, Canada
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Roger A Dixon
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.,Department of Psychology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
81
|
Perakakis N, Yazdani A, Karniadakis GE, Mantzoros C. Omics, big data and machine learning as tools to propel understanding of biological mechanisms and to discover novel diagnostics and therapeutics. Metabolism 2018; 87:A1-A9. [PMID: 30098323 PMCID: PMC6325641 DOI: 10.1016/j.metabol.2018.08.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Nikolaos Perakakis
- Department of Endocrinology, VA Boston Healthcare System, Jamaica Plain, Boston, MA 02130, USA; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alireza Yazdani
- Division of Applied Mathematics, Brown University, Providence, RI 02906, USA
| | | | - Christos Mantzoros
- Department of Endocrinology, VA Boston Healthcare System, Jamaica Plain, Boston, MA 02130, USA; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
82
|
Dienel GA. Metabolomic Assays of Postmortem Brain Extracts: Pitfalls in Extrapolation of Concentrations of Glucose and Amino Acids to Metabolic Dysregulation In Vivo in Neurological Diseases. Neurochem Res 2018; 44:2239-2260. [DOI: 10.1007/s11064-018-2611-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 01/03/2023]
|
83
|
Bagatini MD, dos Santos AA, Cardoso AM, Mânica A, Reschke CR, Carvalho FB. The Impact of Purinergic System Enzymes on Noncommunicable, Neurological, and Degenerative Diseases. J Immunol Res 2018; 2018:4892473. [PMID: 30159340 PMCID: PMC6109496 DOI: 10.1155/2018/4892473] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 07/03/2018] [Accepted: 07/22/2018] [Indexed: 12/11/2022] Open
Abstract
Evidences show that purinergic signaling is involved in processes associated with health and disease, including noncommunicable, neurological, and degenerative diseases. These diseases strike from children to elderly and are generally characterized by progressive deterioration of cells, eventually leading to tissue or organ degeneration. These pathological conditions can be associated with disturbance in the signaling mediated by nucleotides and nucleosides of adenine, in expression or activity of extracellular ectonucleotidases and in activation of P2X and P2Y receptors. Among the best known of these diseases are atherosclerosis, hypertension, cancer, epilepsy, Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). The currently available treatments present limited effectiveness and are mostly palliative. This review aims to present the role of purinergic signaling highlighting the ectonucleotidases E-NTPDase, E-NPP, E-5'-nucleotidase, and adenosine deaminase in noncommunicable, neurological, and degenerative diseases associated with the cardiovascular and central nervous systems and cancer. In conclusion, changes in the activity of ectonucleotidases were verified in all reviewed diseases. Although the role of ectonucleotidases still remains to be further investigated, evidences reviewed here can contribute to a better understanding of the molecular mechanisms of highly complex diseases, which majorly impact on patients' quality of life.
Collapse
Affiliation(s)
- Margarete Dulce Bagatini
- Coordenação Acadêmica, Universidade Federal da Fronteira Sul, Campus Chapecó, Chapecó, SC, Brazil
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | - Andréia Machado Cardoso
- Coordenação Acadêmica, Universidade Federal da Fronteira Sul, Campus Chapecó, Chapecó, SC, Brazil
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Aline Mânica
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Cristina Ruedell Reschke
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Fabiano Barbosa Carvalho
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
84
|
Domínguez-González M, Puigpinós M, Jové M, Naudi A, Portero-Otín M, Pamplona R, Ferrer I. Regional vulnerability to lipoxidative damage and inflammation in normal human brain aging. Exp Gerontol 2018; 111:218-228. [PMID: 30077575 DOI: 10.1016/j.exger.2018.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/29/2018] [Accepted: 07/30/2018] [Indexed: 01/08/2023]
Abstract
Oxidative damage and inflammation coexist in healthy human brain aging. The present study analyzes levels of protein adduction by lipid peroxidation (LP) end-products neuroketal (NKT) and malondialdehyde (MDA), as markers of protein oxidative damage, cycloxygenase-2 (COX-2) levels, as a marker of inflammation, and cytochrome P450 2J2 (CYP2J2), responsible of generation of neuroprotective products, in twelve brain regions in normal middle-aged individuals (MA) and old-aged (OA) individuals. In addition, levels of these markers were evaluated as a function of age as a continuous variable and correction for multiple comparisons. Selection of regions was based on their different vulnerability to prevalent neurodegenerative diseases in aging. Our findings show region-dependent LP end-products, COX-2 and CYP2J2 changes in the aging human brain. However, no clear relationship can be established between NKT, MDA, COX-2 and CYP2J2 levels, and regional vulnerability to neurodegeneration in old age.
Collapse
Affiliation(s)
- Mayelín Domínguez-González
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain
| | - Meritxell Puigpinós
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Alba Naudi
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Manuel Portero-Otín
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain; Biomedical Research Network Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain; Institute of Neurosciences, University of Barcelona, Hospitalet de Llobregat, Spain.
| |
Collapse
|
85
|
Sun J, Jiao C, Ma Y, Chen J, Wu W, Liu S. Anti-ageing effect of red ginseng revealed by urinary metabonomics using RRLC-Q-TOF-MS. PHYTOCHEMICAL ANALYSIS : PCA 2018; 29:387-397. [PMID: 29573298 DOI: 10.1002/pca.2758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/16/2018] [Accepted: 01/30/2018] [Indexed: 06/08/2023]
Abstract
INTRODUCTION Red ginseng (RG) is one of the main processed products of the roots and rhizomes of Panax ginseng C.A. Meyer and is used for anti-ageing. But how metabonomic influences of RG on the progress of ageing are less researched. OBJECTIVE A metabonomic method was developed to study the characters of the ageing process and the effects of total ginsenosides of red ginseng (TGRG) on the progress of ageing. METHODS Urine samples from four different ages (4, 12, 18 and 24 months old) of rats and interference after TGRG were analysed by rapid resolution liquid chromatography coupled with quadruple-time-of-flight mass spectrometry (RRLC-Q-TOF-MS) and multivariate statistical analysis were performed for the pattern recognition and characteristic metabolites identification. RESULTS Fourteen potential biomarkers were found and identified by MS/MS analysis by referring to authentic chemicals. The analysis of metabolic pathways indicated that the reduced energy and lipid metabolism, the decline of kidney function and amino acids metabolism disorders were the main features of ageing. After TGRG administration, lipid and amino acids metabolism of 18 and 24 month-old rats were adjusted to restore a younger level, and nine related biomarkers in the ageing process reset to a younger level were recognised. CONCLUSION These changes showed that TGRG may produce an anti-ageing effect by intervening in the lipid metabolism and correcting the amino acid metabolism disorders in ageing rats.
Collapse
Affiliation(s)
- Jinghui Sun
- College of Pharmacy, Beihua University, Jilin, P. R. China
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Chuanxin Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Yue Ma
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Jianguang Chen
- College of Pharmacy, Beihua University, Jilin, P. R. China
| | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Shuying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| |
Collapse
|
86
|
Chen JJ. Functional MRI of brain physiology in aging and neurodegenerative diseases. Neuroimage 2018; 187:209-225. [PMID: 29793062 DOI: 10.1016/j.neuroimage.2018.05.050] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 05/16/2018] [Accepted: 05/20/2018] [Indexed: 12/14/2022] Open
Abstract
Brain aging and associated neurodegeneration constitute a major societal challenge as well as one for the neuroimaging community. A full understanding of the physiological mechanisms underlying neurodegeneration still eludes medical researchers, fuelling the development of in vivo neuroimaging markers. Hence it is increasingly recognized that our understanding of neurodegenerative processes likely will depend upon the available information provided by imaging techniques. At the same time, the imaging techniques are often developed in response to the desire to observe certain physiological processes. In this context, functional MRI (fMRI), which has for decades provided information on neuronal activity, has evolved into a large family of techniques well suited for in vivo observations of brain physiology. Given the rapid technical advances in fMRI in recent years, this review aims to summarize the physiological basis of fMRI observations in healthy aging as well as in age-related neurodegeneration. This review focuses on in-vivo human brain imaging studies in this review and on disease features that can be imaged using fMRI methods. In addition to providing detailed literature summaries, this review also discusses future directions in the study of brain physiology using fMRI in the clinical setting.
Collapse
Affiliation(s)
- J Jean Chen
- Rotman Research Institute at Baycrest Centre, Canada; Department of Medical Biophysics, University of Toronto, Canada.
| |
Collapse
|
87
|
Xiao Y, Dong J, Yin Z, Wu Q, Zhou Y, Zhou X. Procyanidin B2 protects against d-galactose-induced mimetic aging in mice: Metabolites and microbiome analysis. Food Chem Toxicol 2018; 119:141-149. [PMID: 29751077 DOI: 10.1016/j.fct.2018.05.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/16/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022]
Abstract
To elucidate the possible mechanisms for the preventive effect of procyanidin B2 on aging, a combined analysis of metabolic profile and gut microbiome was carried out in the present study. The mimetic aged mice induced by d-galactose injection (500 mg/kg, sc daily), and the preventive group was fed with the diet plus 0.2% procyanidin B2. After 7 weeks of treatment, the spatial memory was assayed using the Morris water maze test. Procyanidin B2 significantly ameliorated the impaired memory and antioxidant abilities induced by d-galactose. Furthermore, metabolomics analysis of plasma based on LC/Q-TOF-MS demonstrated that phosphatidyl cholines, oleic acid, linoleic acid, carnitine, pantothenic acid, and taurocholic acid were significantly increased in the mice treated with procyanidin B2, and pyruvic acid, hydroxybutyric acid, hippuric acid, and cholic acid were decreased significantly. Together, gut microbiome analysis using Illumina sequencing showed that there were significant differences in the Firmicutes/Bacteroidetes ratio and abundance of Roseburia, Lachnospiraceae, and Bifidobacterium between the aging and supplemental procyanidin B2 groups. In summary, procyanidin B2 possessed potential prevention of the cognitive and oxidative impairment via the metabolic pathway regulation related to citrate cycle, fatty acid, and bile acid in the aged mice, accompanied by remodeling the gut flora.
Collapse
Affiliation(s)
- Ying Xiao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China.
| | - Jialin Dong
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Zhiting Yin
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Qiguo Wu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Yiming Zhou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China
| | - Xiaoli Zhou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, 201418, China.
| |
Collapse
|
88
|
Zhou HJ, Zeng CY, Yang TT, Long FY, Kuang X, Du JR. Lentivirus-mediated klotho up-regulation improves aging-related memory deficits and oxidative stress in senescence-accelerated mouse prone-8 mice. Life Sci 2018; 200:56-62. [DOI: 10.1016/j.lfs.2018.03.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/02/2018] [Accepted: 03/12/2018] [Indexed: 12/13/2022]
|
89
|
Geiszler PC, Ugun-Klusek A, Lawler K, Pardon MC, Yuchun D, Bai L, Daykin CA, Auer DP, Bedford L. Dynamic metabolic patterns tracking neurodegeneration and gliosis following 26S proteasome dysfunction in mouse forebrain neurons. Sci Rep 2018; 8:4833. [PMID: 29555943 PMCID: PMC5859111 DOI: 10.1038/s41598-018-23155-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/02/2018] [Indexed: 01/23/2023] Open
Abstract
Metabolite profiling is an important tool that may better capture the multiple features of neurodegeneration. With the considerable parallels between mouse and human metabolism, the use of metabolomics in mouse models with neurodegenerative pathology provides mechanistic insight and ready translation into aspects of human disease. Using 400 MHz nuclear magnetic resonance spectroscopy we have carried out a temporal region-specific investigation of the metabolome of neuron-specific 26S proteasome knockout mice characterised by progressive neurodegeneration and Lewy-like inclusion formation in the forebrain. An early significant decrease in N-acetyl aspartate revealed evidence of neuronal dysfunction before cell death that may be associated with changes in brain neuroenergetics, underpinning the use of this metabolite to track neuronal health. Importantly, we show early and extensive activation of astrocytes and microglia in response to targeted neuronal dysfunction in this context, but only late changes in myo-inositol; the best established glial cell marker in magnetic resonance spectroscopy studies, supporting recent evidence that additional early neuroinflammatory markers are needed. Our results extend the limited understanding of metabolite changes associated with gliosis and provide evidence that changes in glutamate homeostasis and lactate may correlate with astrocyte activation and have biomarker potential for tracking neuroinflammation.
Collapse
Affiliation(s)
- Philippine C Geiszler
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK.,School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Aslihan Ugun-Klusek
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Karen Lawler
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Ding Yuchun
- School of Computing, University of Newcastle, Newcastle, UK
| | - Li Bai
- School of Computer Sciences, University of Nottingham, Nottingham, UK
| | - Clare A Daykin
- School of Pharmacy, University of Nottingham, Nottingham, UK.,Metaboconsult UK, Heanor, Derbyshire, UK
| | - Dorothee P Auer
- Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK. .,Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, UK.
| | - Lynn Bedford
- School of Life Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
90
|
Abstract
Ischemic stroke is a sudden loss of brain function due to the reduction of blood flow. Brain tissues cease to function with subsequent activation of the ischemic cascade. Metabolomics and lipidomics are modern disciplines that characterize the metabolites and lipid components of a biological system, respectively. Because the pathogenesis of ischemic stroke is heterogeneous and multifactorial, it is crucial to establish comprehensive metabolomic and lipidomic approaches to elucidate these alterations in this disease. Fortunately, metabolomic and lipidomic studies have the distinct advantages of identifying tissue/mechanism-specific biomarkers, predicting treatment and clinical outcome, and improving our understanding of the pathophysiologic basis of disease states. Therefore, recent applications of these analytical approaches in the early diagnosis of ischemic stroke were discussed. In addition, the emerging roles of metabolomics and lipidomics on ischemic stroke were summarized, in order to gain new insights into the mechanisms underlying ischemic stroke and in the search for novel metabolite biomarkers and their related pathways.
Collapse
|
91
|
Kovalchuk A, Nersisyan L, Mandal R, Wishart D, Mancini M, Sidransky D, Kolb B, Kovalchuk O. Growth of Malignant Non-CNS Tumors Alters Brain Metabolome. Front Genet 2018. [PMID: 29515623 PMCID: PMC5826252 DOI: 10.3389/fgene.2018.00041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cancer survivors experience numerous treatment side effects that negatively affect their quality of life. Cognitive side effects are especially insidious, as they affect memory, cognition, and learning. Neurocognitive deficits occur prior to cancer treatment, arising even before cancer diagnosis, and we refer to them as "tumor brain." Metabolomics is a new area of research that focuses on metabolome profiles and provides important mechanistic insights into various human diseases, including cancer, neurodegenerative diseases, and aging. Many neurological diseases and conditions affect metabolic processes in the brain. However, the tumor brain metabolome has never been analyzed. In our study we used direct flow injection/mass spectrometry (DI-MS) analysis to establish the effects of the growth of lung cancer, pancreatic cancer, and sarcoma on the brain metabolome of TumorGraft™ mice. We found that the growth of malignant non-CNS tumors impacted metabolic processes in the brain, affecting protein biosynthesis, and amino acid and sphingolipid metabolism. The observed metabolic changes were similar to those reported for neurodegenerative diseases and brain aging, and may have potential mechanistic value for future analysis of the tumor brain phenomenon.
Collapse
Affiliation(s)
- Anna Kovalchuk
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.,Leaders in Medicine Program, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lilit Nersisyan
- Group of Bioinformatics, Institute of Molecular Biology, National Academy of Sciences, Yerevan, Armenia
| | - Rupasri Mandal
- The Metabolomics Innovation Center, Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - David Wishart
- The Metabolomics Innovation Center, Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Maria Mancini
- Department of Oncology, Champions Oncology, Baltimore, MD, United States
| | - David Sidransky
- Department of Oncology, Champions Oncology, Baltimore, MD, United States.,Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD, United States
| | - Bryan Kolb
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
92
|
Choi JG, Kim SY, Jeong M, Oh MS. Pharmacotherapeutic potential of ginger and its compounds in age-related neurological disorders. Pharmacol Ther 2018; 182:56-69. [PMID: 28842272 DOI: 10.1016/j.pharmthera.2017.08.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Age-related neurological disorders (ANDs), including neurodegenerative diseases, are multifactorial disorders with a risk that increases with aging. ANDs are generally characterized by common neuropathological conditions of the central nervous system, such as oxidative stress, neuroinflammation, and protein misfolding. Recently, efforts have been made to overcome ANDs because of the increase in age-dependent prevalence. Ginger, the rhizome of Zingiber officinale Roscoe, is a popular food spice and has a long history of use in traditional medicine for treating various disease symptoms. The structure-activity relationships of ginger phytochemicals show that ginger can be used to treat ANDs by targeting different ligand sites. This review shows that ginger and its constituents, such as 6-gingerol, 6-shogaol, 6-paradol, zingerone, and dehydrozingerone, are effective for ameliorating the neurological symptoms and pathological conditions of ANDs through by modulating cell death or cell survival signaling molecules. From this review, we conclude that the active ingredients in ginger have therapeutic potential in ANDs.
Collapse
Affiliation(s)
- Jin Gyu Choi
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea
| | - Minsun Jeong
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
93
|
Abstract
Metabolomics, the characterization of metabolites and their changes within biological systems, has seen great technological and methodological progress over the past decade. Most metabolomic experiments involve the characterization of the small-molecule content of fluids or tissue homogenates. While these microliter and larger volume metabolomic measurements can characterize hundreds to thousands of compounds, the coverage of molecular content decreases as sample sizes are reduced to the nanoliter and even to the picoliter volume range. Recent progress has enabled the ability to characterize the major molecules found within specific individual cells. Especially within the brain, a myriad of cell types are colocalized, and oftentimes only a subset of these cells undergo changes in both healthy and pathological states. Here we highlight recent progress in mass spectrometry-based approaches used for single cell metabolomics, emphasizing their application to neuroscience research. Single cell studies can be directed to measuring differences between members of populations of similar cells (e.g., oligodendrocytes), as well as characterizing differences between cell types (e.g., neurons and astrocytes), and are especially useful for measuring changes occurring during different behavior states, exposure to diets and drugs, neuronal activity, and disease. When combined with other omics approaches such as transcriptomics, and with morphological and physiological measurements, single cell metabolomics aids fundamental neurochemical studies, has great potential in pharmaceutical development, and should improve the diagnosis and treatment of brain diseases.
Collapse
Affiliation(s)
- Meng Qi
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Marina C Philip
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Ning Yang
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| |
Collapse
|
94
|
Jové M, Gatius S, Yeramian A, Portero-Otin M, Eritja N, Santacana M, Colas E, Ruiz M, Pamplona R, Matias-Guiu X. Metabotyping human endometrioid endometrial adenocarcinoma reveals an implication of endocannabinoid metabolism. Oncotarget 2018; 7:52364-52374. [PMID: 27429042 PMCID: PMC5239558 DOI: 10.18632/oncotarget.10564] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022] Open
Abstract
Metabolomics, an essential technique in precision medicine, contributes to the molecular fingerprinting of tumours, further helping to understand their pathogenesis. In this work, using a LC-ESI-QTOF-MS/MS platform, we demonstrated the existence of a specific metabolomic signature which could define endometrioid endometrial carcinoma (EEC), arising the endocannabinoid system as a potential pathway involved in EC pathogenesis. Metabolomics could also shed light in the processes involved in myometrial invasion, proposing new targets for possible therapeutic intervention. Consequently, we also described a different metabolomic profile in surface endometrioid carcinoma and myometrial invasive front. We validated pathways disclosed by metabolomics by immunohistochemistry. Specifically, endocannabinoid and purine metabolism could be involved in tumor myometrial invasion.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Sònia Gatius
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Andree Yeramian
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Manuel Portero-Otin
- Department of Experimental Medicine, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Núria Eritja
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Maria Santacana
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Eva Colas
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Maria Ruiz
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, E-25198, Lleida, Spain
| |
Collapse
|
95
|
Convergent synthesis and characterization of fatty acid-conjugated poly(ethylene glycol)-block-poly(epsilon-caprolactone) nanoparticles for improved drug delivery to the brain. Eur Polym J 2018. [DOI: 10.1016/j.eurpolymj.2017.11.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
96
|
Abstract
Multiple diseases have a strong metabolic component, and metabolomics as a powerful phenotyping technology, in combination with orthogonal biological and clinical approaches, will undoubtedly play a determinant role in accelerating the understanding of mechanisms that underlie these complex diseases determined by a set of genetic, lifestyle, and environmental exposure factors. Here, we provide several examples of valuable findings from metabolomics-led studies in diabetes and obesity metabolism, neurodegenerative disorders, and cancer metabolism and offer a longer term vision toward personalized approach to medicine, from population-based studies to pharmacometabolomics.
Collapse
Affiliation(s)
- Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 19, 1005, Lausanne, Switzerland.
| | - Aurelien Thomas
- Unit of Toxicology, CURML, CHUV Lausanne University Hospital, HUG Geneva University Hospitals, Vulliette 04, 1000, Lausanne, Switzerland.
- Faculty of Biology and Medicine, University of Lausanne, Vulliette 04, 1000, Lausanne, Switzerland.
| |
Collapse
|
97
|
Ferrer I. Sisyphus in Neverland. J Alzheimers Dis 2018; 62:1023-1047. [PMID: 29154280 PMCID: PMC5870014 DOI: 10.3233/jad-170609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2017] [Indexed: 11/24/2022]
Abstract
The study of life and living organisms and the way in which these interact and organize to form social communities have been central to my career. I have been fascinated by biology, neurology, and neuropathology, but also by history, sociology, and art. Certain current historical, political, and social events, some occurring proximally but others affecting people in apparently distant places, have had an impact on me. Epicurus, Seneca, and Camus shared their philosophical positions which I learned from. Many scientists from various disciplines have been exciting sources of knowledge as well. I have created a world of hypothesis and experiments but I have also got carried away by serendipity following unexpected observations. It has not been an easy path; errors and wanderings are not uncommon, and opponents close to home much more abundant than one might imagine. Ambition, imagination, resilience, and endurance have been useful in moving ahead in response to setbacks. In the end, I have enjoyed my dedication to science and I am grateful to have glimpsed beauty in it. These are brief memories of a Spanish neuropathologist born and raised in Barcelona, EU.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Service of Pathological Anatomy, Bellvitge University Hospital; CIBERNED; Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
98
|
Metabolomic Estimation of the Diagnosis and Onset Time of Permanent and Transient Cerebral Ischemia. Mol Neurobiol 2017; 55:6193-6200. [PMID: 29270918 DOI: 10.1007/s12035-017-0827-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 12/06/2017] [Indexed: 01/27/2023]
Abstract
Determining the time of stroke onset in order to apply recanalization therapies within the accepted therapeutic window and the correct diagnosis of transient ischemic attack (TIA) are two common clinical problems in acute cerebral ischemia management. Therefore, biomarkers helping in this conundrum could be very helpful. We developed mouse models of distal middle cerebral artery occlusion mimicking TIA and ischemic stroke (IS), respectively. Plasma samples were analyzed by metabolomics at 6, 12, 24, and 48 h post onset in order to find TIA- and time-related stroke biomarkers. The results were validated in a second experimental cohort. Plasma metabolomic profiles identified time after stroke events with a very high accuracy. Specific metabolites pointing to a recent event (< 6 h) were identified. A multivariate (partial least square discriminant analyses [PLS-DA]) model was also able to separate samples from TIA, IS, and sham events with high accuracy and to obtain specific metabolites for each time point. The combination of mice models of focal ischemia with plasma metabolomics allows the discovery of candidate biomarkers for the diagnosis and estimation of onset time of stroke and TIA diagnosis.
Collapse
|
99
|
Sonntag KC, Ryu WI, Amirault KM, Healy RA, Siegel AJ, McPhie DL, Forester B, Cohen BM. Late-onset Alzheimer's disease is associated with inherent changes in bioenergetics profiles. Sci Rep 2017; 7:14038. [PMID: 29070876 PMCID: PMC5656579 DOI: 10.1038/s41598-017-14420-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/10/2017] [Indexed: 12/11/2022] Open
Abstract
Body-wide changes in bioenergetics, i.e., energy metabolism, occur in normal aging and disturbed bioenergetics may be an important contributing mechanism underlying late-onset Alzheimer's disease (LOAD). We investigated the bioenergetic profiles of fibroblasts from LOAD patients and healthy controls, as a function of age and disease. LOAD cells exhibited an impaired mitochondrial metabolic potential and an abnormal redox potential, associated with reduced nicotinamide adenine dinucleotide metabolism and altered citric acid cycle activity, but not with disease-specific changes in mitochondrial mass, production of reactive oxygen species, transmembrane instability, or DNA deletions. LOAD fibroblasts demonstrated a shift in energy production to glycolysis, despite an inability to increase glucose uptake in response to IGF-1. The increase of glycolysis and the abnormal mitochondrial metabolic potential in LOAD appeared to be inherent, as they were disease- and not age-specific. Our findings support the hypothesis that impairment in multiple interacting components of bioenergetic metabolism may be a key mechanism contributing to the risk and pathophysiology of LOAD.
Collapse
Affiliation(s)
- Kai-C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA.
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA.
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA.
| | - Woo-In Ryu
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Kristopher M Amirault
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Ryan A Healy
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Arthur J Siegel
- Internal Medicine Department, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Donna L McPhie
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Brent Forester
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Mood Disorders Division and Geriatric Psychiatry Research Program, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Bruce M Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| |
Collapse
|
100
|
Jové M, Collado R, Quiles JL, Ramírez-Tortosa MC, Sol J, Ruiz-Sanjuan M, Fernandez M, de la Torre Cabrera C, Ramírez-Tortosa C, Granados-Principal S, Sánchez-Rovira P, Pamplona R. A plasma metabolomic signature discloses human breast cancer. Oncotarget 2017; 8:19522-19533. [PMID: 28076849 PMCID: PMC5386702 DOI: 10.18632/oncotarget.14521] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/26/2016] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Metabolomics is the comprehensive global study of metabolites in biological samples. In this retrospective pilot study we explored whether serum metabolomic profile can discriminate the presence of human breast cancer irrespective of the cancer subtype. METHODS Plasma samples were analyzed from healthy women (n = 20) and patients with breast cancer after diagnosis (n = 91) using a liquid chromatography-mass spectrometry platform. Multivariate statistics and a Random Forest (RF) classifier were used to create a metabolomics panel for the diagnosis of human breast cancer. RESULTS Metabolomics correctly distinguished between breast cancer patients and healthy control subjects. In the RF supervised class prediction analysis comparing breast cancer and healthy control groups, RF accurately classified 100% both samples of the breast cancer patients and healthy controls. So, the class error for both group in and the out-of-bag error were 0. We also found 1269 metabolites with different concentration in plasma from healthy controls and cancer patients; and basing on exact mass, retention time and isotopic distribution we identified 35 metabolites. These metabolites mostly support cell growth by providing energy and building stones for the synthesis of essential biomolecules, and function as signal transduction molecules. The collective results of RF, significance testing, and false discovery rate analysis identified several metabolites that were strongly associated with breast cancer. CONCLUSIONS In breast cancer a metabolomics signature of cancer exists and can be detected in patient plasma irrespectively of the breast cancer type.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain
| | - Ricardo Collado
- Department of Oncology, Medical Oncology Unit, Hospital San Pedro de Alcántara, Cáceres, Official Postgraduate Programme in Nutrition and Food Technology, University of Granada, Spain
| | - José Luís Quiles
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, Department of Physiology, University of Granada, Granada, Spain
| | - Mari-Carmen Ramírez-Tortosa
- Institute of Nutrition and Food Technology "José Mataix", Biomedical Research Center, Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
| | - Joaquim Sol
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain
| | | | | | | | - Cesar Ramírez-Tortosa
- Department of Pathological Anatomy, Hospital of Jaén, Jaén, Spain.,GENYO, Centre for Genomics and Oncological Research (Pfizer / University of Granada / Andalusian Regional Government), PTS Granada, Granada, Spain
| | | | | | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain
| |
Collapse
|