51
|
Baumel BS, Doraiswamy PM, Sabbagh M, Wurtman R. Potential Neuroregenerative and Neuroprotective Effects of Uridine/Choline-Enriched Multinutrient Dietary Intervention for Mild Cognitive Impairment: A Narrative Review. Neurol Ther 2021; 10:43-60. [PMID: 33368017 PMCID: PMC8139993 DOI: 10.1007/s40120-020-00227-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/02/2020] [Indexed: 01/21/2023] Open
Abstract
In mild cognitive impairment (MCI) due to Alzheimer disease (AD), also known as prodromal AD, there is evidence for a pathologic shortage of uridine, choline, and docosahexaenoic acid [DHA]), which are key nutrients needed by the brain. Preclinical and clinical evidence shows the importance of nutrient bioavailability to support the development and maintenance of brain structure and function in MCI and AD. Availability of key nutrients is limited in MCI, creating a distinct nutritional need for uridine, choline, and DHA. Evidence suggests that metabolic derangements associated with ageing and disease-related pathology can affect the body's ability to generate and utilize nutrients. This is reflected in lower levels of nutrients measured in the plasma and brains of individuals with MCI and AD dementia, and progressive loss of cognitive performance. The uridine shortage cannot be corrected by normal diet, making uridine a conditionally essential nutrient in affected individuals. It is also challenging to correct the choline shortfall through diet alone, because brain uptake from the plasma significantly decreases with ageing. There is no strong evidence to support the use of single-agent supplements in the management of MCI due to AD. As uridine and choline work synergistically with DHA to increase phosphatidylcholine formation, there is a compelling rationale to combine these nutrients. A multinutrient enriched with uridine, choline, and DHA developed to support brain function has been evaluated in randomized controlled trials covering a spectrum of dementia from MCI to moderate AD. A randomized controlled trial in subjects with prodromal AD showed that multinutrient intervention slowed brain atrophy and improved some measures of cognition. Based on the available clinical evidence, nutritional intervention should be considered as a part of the approach to the management of individuals with MCI due to AD, including adherence to a healthy, balanced diet, and consideration of evidence-based multinutrient supplements.
Collapse
Affiliation(s)
- Barry S Baumel
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Marwan Sabbagh
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Las Vegas, NV, USA
| | - Richard Wurtman
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
52
|
Jones AC, Pinki F, Stewart GS, Costello DA. Inhibition of Urea Transporter (UT)-B Modulates LPS-Induced Inflammatory Responses in BV2 Microglia and N2a Neuroblastoma Cells. Neurochem Res 2021; 46:1322-1329. [PMID: 33675462 DOI: 10.1007/s11064-021-03283-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/20/2021] [Accepted: 02/23/2021] [Indexed: 11/29/2022]
Abstract
Urea is the major nitrogen-containing product of protein metabolism, and the urea cycle is intrinsically linked to nitric oxide (NO) production via the common substrate L-arginine. Urea accumulates in the brain in neurodegenerative states, including Alzheimer's and Huntington's disease. Urea transporter B (UT-B, SLC14A1) is the primary transport protein for urea in the CNS, identified most abundantly in astrocytes. Moreover, enhanced expression of the Slc14a1 gene has been reported under neurodegenerative conditions. While the role of UT-B in disease pathology remains unclear, UT-B-deficient mice display behavioural impairment coupled with urea accumulation, NO disruption and neuronal loss. Recognising the role of inflammation in neurodegenerative disease pathology, the current short study evaluates the role of UT-B in regulating inflammatory responses. Using the specific inhibitor UTBinh-14, we investigated the impact of UT-B inhibition on LPS-induced changes in BV2 microglia and N2a neuroblastoma cells. We found that UTBinh-14 significantly attenuated LPS-induced production of TNFα and IL-6 from BV2 cells, accompanied by reduced release of NO. While we observed a similar reduction in supernatant concentration of IL-6 from N2a cells, the LPS-stimulated NO release was further augmented by UTBinh-14. These changes were accompanied by a small, but significant downregulation in UT-B expression in both cell types following incubation with LPS, which was not restored by UTBinh-14. Taken together, the current evidence implicates UT-B in regulation of inflammatory responses in microglia and neuronal-like cells. Moreover, our findings offer support for the further investigation of UT-B as a novel therapeutic target for neuroinflammatory conditions.
Collapse
Affiliation(s)
- Aimée C Jones
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Farhana Pinki
- UCD School of Biology & Environmental Science, University College Dublin, Dublin 4, Ireland
| | - Gavin S Stewart
- UCD School of Biology & Environmental Science, University College Dublin, Dublin 4, Ireland
| | - Derek A Costello
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland.
- UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
53
|
Erdogan MA, Yigitturk G, Erbas O, Taskıran D. Neuroprotective effects of dexpanthenol on streptozotocin-induced neuronal damage in rats. Drug Chem Toxicol 2021; 45:2160-2168. [PMID: 33874839 DOI: 10.1080/01480545.2021.1914464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
AIM Although the most common age-related neurodegenerative disease defined by memory loss is Alzheimer's disease (AD), only symptomatic therapies are present. A complex pathway for the AD pathogenesis that includes an increase in inflammation has recently been suggested. Since in previous animal experiments dexpanthenol has anti-inflammatory and neuroprotective activities, effects and role of dexpanthenol in an intracerebroventricular (ICV)-streptozotocin (STZ) induced sporadic-AD(memory impairment) animal model have been examined. DESIGN AND METHODS In total, 18 adult sprague-dawley rats were classified into 3 groups; control (n = 6), STZ + Saline (n = 6) and STZ + Dexpanthenol (n = 6). Twelve AD-induced rats through STZ-injection (3 mg/kg) into both lateral ventricles via stereotaxy were separated into two groups five days after STZ administration: one of these groups was treated with dexpanthenol (1000 mg/kg/day, i.p.) for 3 weeks and the other with saline. A passive avoidance learning (PAL) test was used after treatment, followed by brain tissue extraction in all subjects. Brain levels of tumor necrosis factor-alpha (TNF-α) and choline acetyl transferase (ChAT) were measured and Cresyl violet staining was used to count neurons in cornu ammonis-1 (CA1) and cornu ammonis-3 (CA3). RESULTS It was observed that ICV-STZ significantly shortened PAL latency, increased levels of TNF-α in brain, decreased activity of ChAT in brain, and number of hippocampal neurons. However, dexpanthenol significantly reduced all of those STZ-induced harmful effects. CONCLUSION Dexpanthenol significantly prevented the memory deficit induced by ICV-STZ through mitigating neuronal loss in hippocampus, cholinergic deficiency and neuroinflammation in rats. These findings suggest that dexpanthenol may be beneficial for treating memory impairment.
Collapse
Affiliation(s)
- Mumin Alper Erdogan
- Department of Physiology, Faculty of Medicine, Izmir Kâtip Çelebi University, Izmir, Turkey
| | - Gurkan Yigitturk
- Department of Histology, Faculty of Medicine, Mugla University, Mugla, Turkey
| | - Oytun Erbas
- Department of Physiology, Faculty of Medicine, Bilim University, Istanbul, Turkey
| | - Dilek Taskıran
- Department of Physiology, Faculty of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
54
|
Dejakaisaya H, Harutyunyan A, Kwan P, Jones NC. Altered metabolic pathways in a transgenic mouse model suggest mechanistic role of amyloid precursor protein overexpression in Alzheimer's disease. Metabolomics 2021; 17:42. [PMID: 33876332 DOI: 10.1007/s11306-021-01793-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/11/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The mechanistic role of amyloid precursor protein (APP) in Alzheimer's disease (AD) remains unclear. OBJECTIVES Here, we aimed to identify alterations in cerebral metabolites and metabolic pathways in cortex, hippocampus and serum samples from Tg2576 mice, a widely used mouse model of AD. METHODS Metabolomic profilings using liquid chromatography-mass spectrometry were performed and analysed with MetaboAnalyst and weighted correlation network analysis (WGCNA). RESULTS Expressions of 11 metabolites in cortex, including hydroxyphenyllactate-linked to oxidative stress-and phosphatidylserine-lipid metabolism-were significantly different between Tg2576 and WT mice (false discovery rate < 0.05). Four metabolic pathways from cortex, including glycerophospholipid metabolism and pyrimidine metabolism, and one pathway (sulphur metabolism) from hippocampus, were significantly enriched in Tg2576 mice. Network analysis identified five pathways, including alanine, aspartate and glutamate metabolism, and mitochondria electron transport chain, that were significantly correlated with AD genotype. CONCLUSIONS Changes in metabolite concentrations and metabolic pathways are present in the early stage of APP pathology, and may be important for AD development and progression.
Collapse
Affiliation(s)
- Hattapark Dejakaisaya
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, VIC, 3004, Australia
| | - Anna Harutyunyan
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, 3052, Australia
| | - Patrick Kwan
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, VIC, 3004, Australia.
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, 3052, Australia.
| | - Nigel C Jones
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, VIC, 3004, Australia.
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
55
|
Polis B, Karasik D, Samson AO. Alzheimer's disease as a chronic maladaptive polyamine stress response. Aging (Albany NY) 2021; 13:10770-10795. [PMID: 33811757 PMCID: PMC8064158 DOI: 10.18632/aging.202928] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/27/2021] [Indexed: 12/21/2022]
Abstract
Polyamines are nitrogen-rich polycationic ubiquitous bioactive molecules with diverse evolutionary-conserved functions. Their activity interferes with numerous genes' expression resulting in cell proliferation and signaling modulation. The intracellular levels of polyamines are precisely controlled by an evolutionary-conserved machinery. Their transient synthesis is induced by heat stress, radiation, and other traumatic stimuli in a process termed the polyamine stress response (PSR). Notably, polyamine levels decline gradually with age; and external supplementation improves lifespan in model organisms. This corresponds to cytoprotective and reactive oxygen species scavenging properties of polyamines. Paradoxically, age-associated neurodegenerative disorders are characterized by upsurge in polyamines levels, indicating polyamine pleiotropic, adaptive, and pathogenic roles. Specifically, arginase overactivation and arginine brain deprivation have been shown to play an important role in Alzheimer's disease (AD) pathogenesis. Here, we assert that a universal short-term PSR associated with acute stimuli is beneficial for survival. However, it becomes detrimental and maladaptive following chronic noxious stimuli, especially in an aging organism. Furthermore, we regard cellular senescence as an adaptive response to stress and suggest that PSR plays a central role in age-related neurodegenerative diseases' pathogenesis. Our perspective on AD proposes an inclusive reassessment of the causal relationships between the classical hallmarks and clinical manifestation. Consequently, we offer a novel treatment strategy predicated upon this view and suggest fine-tuning of arginase activity with natural inhibitors to preclude or halt the development of AD-related dementia.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - David Karasik
- Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA 02131, USA
- Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Abraham O. Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
56
|
Widespread severe cerebral elevations of haptoglobin and haemopexin in sporadic Alzheimer's disease: Evidence for a pervasive microvasculopathy. Biochem Biophys Res Commun 2021; 555:89-94. [PMID: 33813281 DOI: 10.1016/j.bbrc.2021.02.107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022]
Abstract
Sporadic Alzheimer's disease (sAD) is the commonest cause of age-related neurodegeneration but there are no available treatments with demonstrated disease-modifying actions. It is therefore relevant to study hitherto-unknown aspects of brain structure and function to seek new disease-related mechanisms that might be targeted by novel disease-modifying interventions. During hypothesis-generating proteomic investigations in a case-control study of sAD, we observed widespread elevations of haptoglobin and haemopexin in all six brain-regions studied, which together represent much of the brain. Measured perturbations were significant, with the posterior probability of upregulation generally >95% and haptoglobin doubling in expression levels on average across deep brain structures (hippocampus, entorhinal cortex and cingulate gyrus) as well as sensory and motor cortices, and cerebellum. Haptoglobin and haemopexin are often regarded as circulating proteins whose main functions are to bind, respectively, the strongly pro-inflammatory extracellular haemoglobin and haeme molecules that form following haemolysis, thereby promoting their clearance and suppressing damage they might otherwise cause, for example, acute kidney injury. To our knowledge, elevations in neither cerebral haptoglobin nor haemopexin have previously been linked to the pathogenesis of sAD. Post-mortem examination of these cases showed no signs of macroscopic cerebral haemorrhage. These findings demonstrate pervasive cerebral elevation of haptoglobin and haemopexin, consistent with low-level intracerebral leakage of haemoglobin and consequent haeme formation throughout sAD brain. They point to a widespread underlying microvasculopathy that facilitates erythrocyte leakage, thereby triggering elevated tissue-free haemoglobin and driving the measured elevations in haptoglobin and haemopexin.
Collapse
|
57
|
Mechanistic Insights into Alzheimer's Disease Unveiled through the Investigation of Disturbances in Central Metabolites and Metabolic Pathways. Biomedicines 2021; 9:biomedicines9030298. [PMID: 33799385 PMCID: PMC7998757 DOI: 10.3390/biomedicines9030298] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Hydrophilic metabolites are closely involved in multiple primary metabolic pathways and, consequently, play an essential role in the onset and progression of multifactorial human disorders, such as Alzheimer’s disease. This review article provides a comprehensive revision of the literature published on the use of mass spectrometry-based metabolomics platforms for approaching the central metabolome in Alzheimer’s disease research, including direct mass spectrometry, gas chromatography-mass spectrometry, hydrophilic interaction liquid chromatography-mass spectrometry, and capillary electrophoresis-mass spectrometry. Overall, mounting evidence points to profound disturbances that affect a multitude of central metabolic pathways, such as the energy-related metabolism, the urea cycle, the homeostasis of amino acids, fatty acids and nucleotides, neurotransmission, and others.
Collapse
|
58
|
Altiné‐Samey R, Antier D, Mavel S, Dufour‐Rainfray D, Balageas A, Beaufils E, Emond P, Foucault‐Fruchard L, Chalon S. The contributions of metabolomics in the discovery of new therapeutic targets in Alzheimer's disease. Fundam Clin Pharmacol 2021; 35:582-594. [DOI: 10.1111/fcp.12654] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/05/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Affiliation(s)
| | - Daniel Antier
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service Pharmacie Tours France
| | - Sylvie Mavel
- UMR 1253 iBrain Université de Tours Inserm, Tours France
| | - Diane Dufour‐Rainfray
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service de Médecine Nucléaire In Vitro Tours France
| | | | - Emilie Beaufils
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Centre Mémoire Ressources et Recherche Tours France
| | - Patrick Emond
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service de Médecine Nucléaire In Vitro Tours France
| | - Laura Foucault‐Fruchard
- UMR 1253 iBrain Université de Tours Inserm, Tours France
- CHU Tours Service Pharmacie Tours France
| | - Sylvie Chalon
- UMR 1253 iBrain Université de Tours Inserm, Tours France
| |
Collapse
|
59
|
Scholefield M, Church SJ, Xu J, Patassini S, Roncaroli F, Hooper NM, Unwin RD, Cooper GJS. Widespread Decreases in Cerebral Copper Are Common to Parkinson's Disease Dementia and Alzheimer's Disease Dementia. Front Aging Neurosci 2021; 13:641222. [PMID: 33746735 PMCID: PMC7966713 DOI: 10.3389/fnagi.2021.641222] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/05/2021] [Indexed: 01/24/2023] Open
Abstract
Several studies of Parkinson's disease (PD) have reported dysregulation of cerebral metals, particularly decreases in copper and increases in iron in substantia nigra (SN). However, few studies have investigated regions outside the SN, fewer have measured levels of multiple metals across different regions within the same brains, and there are no currently-available reports of metal levels in Parkinson's disease dementia (PDD). This study aimed to compare concentrations of nine essential metals across nine different brain regions in cases of PDD and controls. Investigated were: primary motor cortex (MCX); cingulate gyrus (CG); primary visual cortex (PVC); hippocampus (HP); cerebellar cortex (CB); SN; locus coeruleus (LC); medulla oblongata (MED); and middle temporal gyrus (MTG), thus covering regions with severe, moderate, or low levels of neuronal loss in PDD. Levels of eight essential metals and selenium were determined using an analytical methodology involving the use of inductively-coupled plasma mass spectrometry (ICP-MS), and compared between cases and controls, to better understand the extent and severity of metal perturbations. Findings were also compared with those from our previous study of sporadic Alzheimer's disease dementia (ADD), which employed equivalent methods, to identify differences and similarities between these conditions. Widespread copper decreases occurred in PDD in seven of nine regions (exceptions being LC and CB). Four PDD-affected regions showed similar decreases in ADD: CG, HP, MTG, and MCX. Decreases in potassium and manganese were present in HP, MTG and MCX; decreased manganese was also found in SN and MED. Decreased selenium and magnesium were present in MCX, and decreased zinc in HP. There was no evidence for increased iron in SN or any other region. These results identify alterations in levels of several metals across multiple regions of PDD brain, the commonest being widespread decreases in copper that closely resemble those in ADD, pointing to similar disease mechanisms in both dementias.
Collapse
Affiliation(s)
- Melissa Scholefield
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie J. Church
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jingshu Xu
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stefano Patassini
- Faculty of Science, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Brain and Mental Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Nigel M. Hooper
- Division of Neuroscience & Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Richard D. Unwin
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Stoller Biomarker Discovery Centre & Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester, United Kingdom
| | - Garth J. S. Cooper
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Faculty of Science, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
60
|
Dienel GA. Stop the rot. Enzyme inactivation at brain harvest prevents artifacts: A guide for preservation of the in vivo concentrations of brain constituents. J Neurochem 2021; 158:1007-1031. [PMID: 33636013 DOI: 10.1111/jnc.15293] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 12/25/2022]
Abstract
Post-mortem metabolism is widely recognized to cause rapid and prolonged changes in the concentrations of multiple classes of compounds in brain, that is, they are labile. Post-mortem changes from levels in living brain include components of pathways of metabolism of glucose and energy compounds, amino acids, lipids, signaling molecules, neuropeptides, phosphoproteins, and proteins. Methods that stop enzyme activity at brain harvest were developed almost 50 years ago and have been extensively used in studies of brain functions and diseases. Unfortunately, these methods are not commonly used to harvest brain tissue for mass spectrometry-based metabolomic studies or for imaging mass spectrometry studies (IMS, also called mass spectrometry imaging, MSI, or matrix-assisted laser desorption/ionization-MSI, MALDI-MSI). Instead these studies commonly kill animals, decapitate, dissect out brain and regions of interest if needed, then 'snap' freeze the tissue to stop enzymatic activity after harvest, with post-mortem intervals typically ranging from ~0.5 to 3 min. To increase awareness of the importance of stopping metabolism at harvest and preventing the unnecessary complications of not doing so, this commentary provides examples of labile metabolites and the magnitudes of their post-mortem changes in concentrations during brain harvest. Brain harvest methods that stop metabolism at harvest eliminate post-mortem enzymatic activities and can improve characterization of normal and diseased brain. In addition, metabolomic studies would be improved by reporting absolute units of concentration along with normalized peak areas or fold changes. Then reported values can be evaluated and compared with the extensive neurochemical literature to help prevent reporting of artifactual data.
Collapse
Affiliation(s)
- Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
61
|
Terburgh K, Coetzer J, Lindeque JZ, van der Westhuizen FH, Louw R. Aberrant BCAA and glutamate metabolism linked to regional neurodegeneration in a mouse model of Leigh syndrome. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166082. [PMID: 33486097 DOI: 10.1016/j.bbadis.2021.166082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/23/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
The dysfunction of respiratory chain complex I (CI) is the most common form of mitochondrial disease that most often presents as Leigh syndrome (LS) in children - a severe neurometabolic disorder defined by progressive focal lesions in specific brain regions. The mechanisms underlying this region-specific vulnerability to CI deficiency, however, remain elusive. Here, we examined brain regional respiratory chain enzyme activities and metabolic profiles in a mouse model of LS with global CI deficiency to gain insight into regional vulnerability to neurodegeneration. One lesion-resistant and three lesion-prone brain regions were investigated in Ndufs4 knockout (KO) mice at the late stage of LS. Enzyme assays confirmed significantly decreased (60-80%) CI activity in all investigated KO brain regions, with the lesion-resistant region displaying the highest residual CI activity (38% of wild type). A higher residual CI activity, and a less perturbed NADH/NAD+ ratio, correlate with less severe metabolic perturbations in KO brain regions. Moreover, less perturbed BCAA oxidation and increased glutamate oxidation seem to distinguish lesion-resistant from -prone KO brain regions, thereby identifying key areas of metabolism to target in future therapeutic intervention studies.
Collapse
Affiliation(s)
- Karin Terburgh
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), South Africa
| | - Janeé Coetzer
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), South Africa
| | - Jeremy Z Lindeque
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), South Africa
| | - Francois H van der Westhuizen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), South Africa.
| |
Collapse
|
62
|
Design, synthesis, and biological activity of novel semicarbazones as potent Ryanodine receptor1 inhibitors of Alzheimer’s disease. Bioorg Med Chem 2021; 29:115891. [DOI: 10.1016/j.bmc.2020.115891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023]
|
63
|
Kinoshita K, Otsuka R, Takada M, Tsukamoto-Yasui M, Nishita Y, Tange C, Tomida M, Shimokata H, Kuzuya M, Imaizumi A, Arai H. The Association between Dietary Amino Acid Intake and Cognitive Decline 8 Years Later in Japanese Community-Dwelling Older Adults. J Nutr Health Aging 2021; 25:165-171. [PMID: 33491030 DOI: 10.1007/s12603-020-1470-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Previous studies have reported a relationship between low protein intake and cognitive decline and have suggested that this association may be related to specific amino acid intake. However, the effects of amino acid intake on the maintenance of cognitive function have yet to be clarified. We examined the longitudinal association between dietary amino acid intake and cognitive function in community-dwelling older adults. DESIGN Longitudinal epidemiological study. SETTING Community-based setting. PARTICIPANTS This study comprised 427 study participants aged 60-82 years with no cognitive decline, defined as a Mini-Mental State Examination (MMSE) score of >27 at baseline, who also participated in a follow-up. The average and standard deviation of the follow-up period was 8.2 ± 0.3 years. MEASUREMENTS Dietary intake was assessed using three-day dietary records at baseline. Participants were classified into quartiles (Q1-Q4) based on the intake of 19 amino acids for males and females. Next, we classified participants into Q1 and Q2-Q4 groups. Cognitive function was assessed using the MMSE both at baseline and at follow-up. Multivariable logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for the associations between the Q1 group and cognitive decline (MMSE ≤27), using the Q2-Q4 group as a reference group. Covariates were age, sex, body mass index, years of education, severity of depressive symptoms, history of lifestyle diseases (hypertension, dyslipidemia, diabetes mellitus, stroke, and ischemic heart disease), energy intake (kcal/d), protein intake (g/d), and MMSE score at baseline. RESULTS Cognitive decline was present in 133 (31.1%) participants. After adjustment for covariates, including total protein intake, the ORs (95% CIs) for cognitive decline were 2.40 (1.21-4.75) for lysine, 2.05 (1.02-4.09) for phenylalanine, 2.18 (1.09-4.34) for threonine, and 2.10 (1.06-4.15) for alanine. CONCLUSION The results suggest that lysine, phenylalanine, threonine, and alanine intake is important for the maintenance of cognitive function in older people, independent of total protein intake.
Collapse
Affiliation(s)
- K Kinoshita
- Rei Otsuka, Section of NILS-LSA, Center for Gerontology and Social Science, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi 474-8511, Japan, Tel: +81-562-46-2311; FAX: +81-562-46-2373; E-mail:
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Wu ZE, Fraser K, Kruger MC, Sequeira IR, Yip W, Lu LW, Plank LD, Murphy R, Cooper GJS, Martin JC, Poppitt SD. Metabolomic signatures for visceral adiposity and dysglycaemia in Asian Chinese and Caucasian European adults: the cross-sectional TOFI_Asia study. Nutr Metab (Lond) 2020; 17:95. [PMID: 33292338 PMCID: PMC7667766 DOI: 10.1186/s12986-020-00518-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Asian Chinese are more susceptible to deposition of visceral adipose tissue (VAT) and type 2 diabetes (T2D) development than European Caucasians when matched for gender, age and body mass index (BMI). Our aims were: (i) characterise the ethnicity-specific metabolomic signature of visceral adiposity measured by dual energy X-ray absorptiometry (DXA) and fasting plasma glucose (FPG), and (ii) identify individuals susceptible to worse metabolic health outcomes. METHODS Fasting plasma samples from normoglycaemic (n = 274) and prediabetic (n = 83) participants were analysed with liquid chromatography-mass spectrometry using untargeted metabolomics. Multiple linear regression adjusting for age, gender and BMI was performed to identify metabolites associated with FPG and VAT calculated as percentage of total body fat (%VATTBF) in each ethnic group. Metabolic risk groups in each ethnicity were stratified based on the joint metabolomic signature for FPG and %VATTBF and clinically characterised using partial least squares-discriminant analysis (PLS-DA) and t-tests. RESULTS FPG was correlated with 40 and 110 metabolites in Caucasians and Chinese respectively, with diglyceride DG(38:5) (adjusted β = 0.29, p = 3.00E-05) in Caucasians and triglyceride TG(54:4) (adjusted β = 0.28, p = 2.02E-07) in Chinese being the most significantly correlated metabolite based on the p-value. %VATTBF was correlated with 85 and 119 metabolites in Caucasians and Chinese respectively, with TG(56:2) (adjusted β = 0.3, p = 8.25E-09) in Caucasians and TG(58:3) (adjusted β = 0.25, p = 2.34E-08) in Chinese being the most significantly correlated. 24 metabolites associated with FPG were common to both ethnicities including glycerolipid species. 67 metabolites associated with %VATTBF were common to both ethnicities including positive correlations with dihydroceramide, sphingomyelin, glycerolipid, phosphatidylcholine, phosphatidylethnolamine, and inverse correlations with ether-linked phosphatidylcholine. Participant re-stratification found greater total and central adiposity, worse clinical lipid profiles, higher serum glucoregulatory peptides and liver enzymes in normal fasting glucose (NFG) individuals with a prediabetic metabolomic profile than NFG individuals with a normoglycaemic metabolomic profile in both ethnicities. CONCLUSIONS Untargeted metabolomics identified common and disparate metabolites associated with FPG and %VATTBF, with an ethnic-dimorphic signature for these metabolic traits. These signatures could improve risk stratification and identify NFG individuals with an adverse cardiometabolic and T2D risk profile.
Collapse
Affiliation(s)
- Zhanxuan E Wu
- Food Nutrition and Health, Food and Bio-Based Products, AgResearch Limited, Palmerston North, 4442, New Zealand.,School of Health Sciences, Massey University, Palmerston North, 4442, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Karl Fraser
- Food Nutrition and Health, Food and Bio-Based Products, AgResearch Limited, Palmerston North, 4442, New Zealand. .,High-Value Nutrition National Science Challenge, Auckland, New Zealand. .,Riddet Institute, Massey University, Palmerston North, 4442, New Zealand.
| | - Marlena C Kruger
- School of Health Sciences, Massey University, Palmerston North, 4442, New Zealand.,Riddet Institute, Massey University, Palmerston North, 4442, New Zealand
| | - Ivana R Sequeira
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Wilson Yip
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Louise W Lu
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Lindsay D Plank
- Department of Surgery, University of Auckland, Auckland, 1010, New Zealand
| | - Rinki Murphy
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Department of Medicine, School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Garth J S Cooper
- Department of Medicine, School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand.,Centre for Advanced Discovery and Experimental Therapeutics, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9NT, UK
| | | | - Sally D Poppitt
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Riddet Institute, Massey University, Palmerston North, 4442, New Zealand.,Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand.,Department of Medicine, School of Biological Sciences, University of Auckland, Auckland, 1010, New Zealand
| |
Collapse
|
65
|
Zhao Y, Chen H, Iqbal J, Liu X, Zhang H, Xiao S, Jin N, Yao F, Shen L. Targeted metabolomics study of early pathological features in hippocampus of triple transgenic Alzheimer's disease male mice. J Neurosci Res 2020; 99:927-946. [PMID: 33197957 DOI: 10.1002/jnr.24750] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a serious neurodegenerative disease in people of age 65 or above. The detailed etiology and pathogenesis of AD have not been elucidated yet. In this study, the hippocampi of 2- and 6-month-old triple transgenic Alzheimer's disease male mice and age-sex-matched wild-type (WT) mice were analyzed by using targeted metabolomics approach. Compared with WT mice, 24 and 60 metabolites were found with significant differences in 2- and 6-month-old AD mice. Among these, 14 metabolites were found common while 10 metabolites showed consistent variable trends in both groups. These differential metabolites are found associated with amino acid, lipid, vitamin, nucleotide-related base, neurotransmitter and energy metabolisms, and oxidative stress. The results suggest that these differential metabolites might play a critical role in AD pathophysiology, and may serve as potential biomarkers for AD. Moreover, the results highlight the involvement of abnormal purine, pyrimidine, arginine, and proline metabolism, along with glycerophospholipid metabolism in early pathology of AD. For the first time, several differential metabolites are found to be associated with AD in this study. Targeted metabolomics can be used for rapid and accurate quantitative analysis of specific target metabolites associated with AD.
Collapse
Affiliation(s)
- Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Haiquan Chen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Javed Iqbal
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China.,Shenzhen Bay Laboratory, Shenzhen, P.R. China
| | - Shifeng Xiao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Na Jin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| | - Fang Yao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China.,Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, P.R. China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P.R. China
| |
Collapse
|
66
|
Scholefield M, Unwin RD, Cooper GJ. Shared perturbations in the metallome and metabolome of Alzheimer's, Parkinson's, Huntington's, and dementia with Lewy bodies: A systematic review. Ageing Res Rev 2020; 63:101152. [PMID: 32846222 DOI: 10.1016/j.arr.2020.101152] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
Despite differences in presentation, age-related dementing diseases such as Alzheimer's (AD), Parkinson's (PD), and Huntington's diseases (HD), and dementia with Lewy bodies (DLB) may share pathogenic processes. This review aims to systematically assemble and compare findings in various biochemical pathways across these four dementias. PubMed and Google Scholar were screened for articles reporting on brain and biofluid measurements of metals and/or metabolites in AD, PD, HD, or DLB. Articles were assessed using specific a priori-defined inclusion and exclusion criteria. Of 284 papers identified, 198 met criteria for inclusion. Although varying coverage levels of metals and metabolites across diseases and tissues made comparison of many analytes impossible, several common findings were identified: elevated glucose in both brain tissue and biofluids of AD, PD, and HD cases; increased iron and decreased copper in AD, PD and HD brain tissue; and decreased uric acid in biofluids of AD and PD cases. Other analytes were found to differ between diseases or were otherwise not covered across all conditions. These findings indicate that disturbances in glucose and purine pathways may be common to AD, PD, and HD. However, standardisation of methodologies and better coverage in some areas - notably of DLB - are necessary to validate and extend these findings.
Collapse
|
67
|
Scholefield M, Church SJ, Xu J, Robinson AC, Gardiner NJ, Roncaroli F, Hooper NM, Unwin RD, Cooper GJS. Effects of Alterations of Post-Mortem Delay and Other Tissue-Collection Variables on Metabolite Levels in Human and Rat Brain. Metabolites 2020; 10:E438. [PMID: 33138273 PMCID: PMC7694048 DOI: 10.3390/metabo10110438] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
The use of post-mortem human tissue is indispensable in studies investigating alterations in metabolite levels in neurodegenerative conditions such as Alzheimer's disease (AD). However, variability between samples may have unknown effects on metabolite concentrations. The aim of this study was to characterize the impact of such variables. Cingulate gyrus was obtained from AD cases and controls, from three brain banks. Gas chromatography-mass spectrometry (GC-MS) was used to measure and compare the levels of 66 identifiable metabolites in these tissues to determine effects of tissue-collection variables. The effect of PMD was further investigated by analysis of rat brain cortex and cerebellum collected following post-mortem delays (PMDs) of zero to 72 h. Metabolite levels between cases and controls were not replicable across cohorts with variable age- and gender-matching, PMD, and control Braak staging. Analysis of rat tissues found significant effects of PMD on 31 of 63 identified metabolites over periods up to 72 h. PMD must be kept under 24 h for metabolomics analyses on brain tissues to yield replicable results. Tissues should also be well age- and gender-matched, and Braak stage in controls should be kept to a minimum in order to minimize the impact of these variables in influencing metabolite variability.
Collapse
Affiliation(s)
- Melissa Scholefield
- Centre for Advanced Discovery & Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M19 9NT, UK; (S.J.C.); (J.X.); (R.D.U.); (G.J.S.C.)
| | - Stephanie J. Church
- Centre for Advanced Discovery & Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M19 9NT, UK; (S.J.C.); (J.X.); (R.D.U.); (G.J.S.C.)
| | - Jingshu Xu
- Centre for Advanced Discovery & Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M19 9NT, UK; (S.J.C.); (J.X.); (R.D.U.); (G.J.S.C.)
- School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag 92 019, Auckland 1142, New Zealand
| | - Andrew C. Robinson
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Salford Royal Hospital, Salford M6 8HD, UK;
| | - Natalie J. Gardiner
- Division of Diabetes, Endocrinology & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Oxford Rd, Manchester M13 9PL, UK;
| | - Federico Roncaroli
- Division of Neuroscience & Experimental Psychology, and Lydia Becker Institute of Immunology & Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M19 9NT, UK;
| | - Nigel M. Hooper
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M19 9NT, UK;
| | - Richard D. Unwin
- Centre for Advanced Discovery & Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M19 9NT, UK; (S.J.C.); (J.X.); (R.D.U.); (G.J.S.C.)
- Stoller Biomarker Discovery Centre & Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Citylabs 1.0 (Third Floor), Nelson Street, Manchester M13 9NQ, UK
| | - Garth J. S. Cooper
- Centre for Advanced Discovery & Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M19 9NT, UK; (S.J.C.); (J.X.); (R.D.U.); (G.J.S.C.)
- School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag 92 019, Auckland 1142, New Zealand
| |
Collapse
|
68
|
Hunsberger HC, Greenwood BP, Tolstikov V, Narain NR, Kiebish MA, Denny CA. Divergence in the metabolome between natural aging and Alzheimer's disease. Sci Rep 2020; 10:12171. [PMID: 32699218 PMCID: PMC7376199 DOI: 10.1038/s41598-020-68739-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 06/18/2020] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive and debilitating neurodegenerative disorder and one of the leading causes of death in the United States. Although amyloid plaques and fibrillary tangles are hallmarks of AD, research suggests that pathology associated with AD often begins 20 or more years before symptoms appear. Therefore, it is essential to identify early-stage biomarkers in those at risk for AD and age-related cognitive decline (ARCD) in order to develop preventative treatments. Here, we used an untargeted metabolomics analysis to define system-level alterations following cognitive decline in aged and APP/PS1 (AD) mice. At 6, 12, and 24 months of age, both control (Ctrl) and AD mice were tested in a 3-shock contextual fear conditioning (CFC) paradigm to assess memory decline. AD mice exhibited memory deficits across age and these memory deficits were also seen in naturally aged mice. Prefrontal cortex (PFC), hippocampus (HPC), and spleen were then collected and analyzed for metabolomic alterations. A number of significant pathways were altered between Ctrl and AD mice and naturally aged mice. By identifying systems-level alterations following ARCD and AD, these data could provide insights into disease mechanisms and advance the development of biomarker panels.
Collapse
Affiliation(s)
- Holly C Hunsberger
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), NYSPI Kolb Research Annex, Room 777, 1051 Riverside Drive, Unit 87, New York, NY, USA
| | | | | | | | | | - Christine Ann Denny
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, USA.
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), NYSPI Kolb Research Annex, Room 777, 1051 Riverside Drive, Unit 87, New York, NY, USA.
| |
Collapse
|
69
|
Agunloye OM, Oboh G. Effect of diet supplemented with P. ostreatus and L. subnudus on memory index and key enzymes linked with Alzheimer's disease in streptozotocin-induced diabetes rats. J Food Biochem 2020; 45:e13355. [PMID: 32628296 DOI: 10.1111/jfbc.13355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/11/2020] [Accepted: 06/06/2020] [Indexed: 12/22/2022]
Abstract
Mushrooms have been reported in folklore for the management of Alzheimer's disease (AD) and diabetes with limited scientific prove. This study aims to unravel the effect of diets supplemented with P. ostreatus and L. subnudus on diabetic encephalopathy (DE) in streptozotocin (STZ)-induced diabetic rats. The memory index, brain cholinesterase, arginase activity, and markers for oxidative stress were evaluated. STZ-induced diabetic rats exhibited memory deficits, elevated brain cholinesterase, arginase activity in comparison with nondiabetic rats. Also, markers for oxidative stress were altered in the brain of diabetic rat when compare with nondiabetic rats. Meanwhile, diabetic rats fed with supplemented-diets exhibited better memory index, reduced cholinesterase, arginase activity in comparison with untreated diabetic rats placed on a basal diet. Also, supplemented-diets restored altered markers of oxidative stress. Our findings indicated that P. ostreatus and L. subnudus-supplemented diets could prevent DE. Nevertheless, diets supplemented with L. subnudus had better nutraceutical potential than P. ostreatus. PRACTICAL APPLICATIONS: Diabetes mellitus is one of the most chronic diseases in the world. Also, it is a risk factor for several complications such as cognitive dysfunction, hypertension, and other health issues. Mushrooms are commonly consumed as food or as food supplements in many countries of the world and are a rich source of protein, chitin, and vitamins. Diet supplemented with P. ostreatus and L. subnudus were able to restore memory deficit in diabetic rats, proven to be a dietary intervention in the management of memory deficit linked with diabetes mellitus. Findings from this study show that consumption of P. ostreatus and L. subnudus as food or supplement could help in a diabetic state.
Collapse
Affiliation(s)
- Odunayo M Agunloye
- Functional Foods, Nutraceuticals and Phytomedicine Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Ganiyu Oboh
- Functional Foods, Nutraceuticals and Phytomedicine Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
70
|
Cabrera D, Kruger M, Wolber FM, Roy NC, Fraser K. Effects of short- and long-term glucocorticoid-induced osteoporosis on plasma metabolome and lipidome of ovariectomized sheep. BMC Musculoskelet Disord 2020; 21:349. [PMID: 32503480 PMCID: PMC7275480 DOI: 10.1186/s12891-020-03362-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/25/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Understanding the metabolic and lipidomic changes that accompany bone loss in osteoporosis might provide insights about the mechanisms behind molecular changes and facilitate developing new drugs or nutritional strategies for osteoporosis prevention. This study aimed to examine the effects of short- or long-term glucocorticoid-induced osteoporosis on plasma metabolites and lipids of ovariectomized (OVX) sheep. METHODS Twenty-eight aged ewes were divided randomly into four groups: an OVX group, OVX in combination with glucocorticoids for two months (OVXG2), and OVX in combination with five doses of glucocorticoids (OVXG5) to induce bone loss, and a control group. Liquid chromatography-mass spectrometry untargeted metabolomic analysis was applied to monthly plasma samples to follow the progression of osteoporosis over five months. RESULTS The metabolite profiles revealed significant differences in the plasma metabolome of OVX sheep and OVXG when compared with the control group by univariate analysis. Nine metabolites were altered, namely 5-methoxytryptophan, valine, methionine, tryptophan, glutaric acid, 2-pyrrolidone-5-carboxylic acid, indole-3-carboxaldehyde, 5-hydroxylysine and malic acid. Similarly, fifteen lipids were perturbed from multiple lipid classes such as lysophoslipids, phospholipids and ceramides. CONCLUSION This study showed that OVX and glucocorticoid interventions altered the metabolite and lipid profiles of sheep, suggesting that amino acid and lipid metabolisms are potentially the main perturbed metabolic pathways regulating bone loss in OVX sheep.
Collapse
Affiliation(s)
- Diana Cabrera
- Food Nutrition & Health Team, AgResearch Grasslands, Tennent Drive, Palmerston North, 4442 New Zealand
| | - Marlena Kruger
- School of Health Sciences, Massey University, Tennent Drive, Palmerston North, 4442 New Zealand
- Riddet Institute, Massey University, Palmerston North, 4442 New Zealand
| | - Frances M. Wolber
- Riddet Institute, Massey University, Palmerston North, 4442 New Zealand
- School of Food Advanced technology, Massey University, Tennent Drive, Palmerston North, 4442 New Zealand
- Centre for Metabolic Health Research, Massey University, Tennent Drive, Palmerston North, 4442 New Zealand
| | - Nicole C. Roy
- Food Nutrition & Health Team, AgResearch Grasslands, Tennent Drive, Palmerston North, 4442 New Zealand
- Riddet Institute, Massey University, Palmerston North, 4442 New Zealand
- High-Value Nutrition National Science Challenge, Auckland, 1142 New Zealand
| | - Karl Fraser
- Food Nutrition & Health Team, AgResearch Grasslands, Tennent Drive, Palmerston North, 4442 New Zealand
- Riddet Institute, Massey University, Palmerston North, 4442 New Zealand
- High-Value Nutrition National Science Challenge, Auckland, 1142 New Zealand
| |
Collapse
|
71
|
Long DM, Frame AK, Reardon PN, Cumming RC, Hendrix DA, Kretzschmar D, Giebultowicz JM. Lactate dehydrogenase expression modulates longevity and neurodegeneration in Drosophila melanogaster. Aging (Albany NY) 2020; 12:10041-10058. [PMID: 32484787 PMCID: PMC7346061 DOI: 10.18632/aging.103373] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/14/2020] [Indexed: 11/25/2022]
Abstract
Lactate dehydrogenase (LDH) catalyzes the conversion of glycolysis-derived pyruvate to lactate. Lactate has been shown to play key roles in brain energetics and memory formation. However, lactate levels are elevated in aging and Alzheimer's disease patients, and it is not clear whether lactate plays protective or detrimental roles in these contexts. Here we show that Ldh transcript levels are elevated and cycle with diurnal rhythm in the heads of aged flies and this is associated with increased LDH protein, enzyme activity, and lactate concentrations. To understand the biological significance of increased Ldh gene expression, we genetically manipulated Ldh levels in adult neurons or glia. Overexpression of Ldh in both cell types caused a significant reduction in lifespan whereas Ldh down-regulation resulted in lifespan extension. Moreover, pan-neuronal overexpression of Ldh disrupted circadian locomotor activity rhythms and significantly increased brain neurodegeneration. In contrast, reduction of Ldh in neurons delayed age-dependent neurodegeneration. Thus, our unbiased genetic approach identified Ldh and lactate as potential modulators of aging and longevity in flies.
Collapse
Affiliation(s)
- Dani M Long
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA.,Present address: Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ariel K Frame
- Department of Biology, Western University of London, London N6A 5B7, Ontario, Canada
| | | | - Robert C Cumming
- Department of Biology, Western University of London, London N6A 5B7, Ontario, Canada
| | - David A Hendrix
- Department of Biochemistry and Biophysics, School of Electrical Engineering and Computer Science, Corvallis, OR 97331, USA
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | | |
Collapse
|
72
|
Anand U, Korchev Y, Anand P. The role of urea in neuronal degeneration and sensitization: An in vitro model of uremic neuropathy. Mol Pain 2020; 15:1744806919881038. [PMID: 31549574 PMCID: PMC6796209 DOI: 10.1177/1744806919881038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Uremic neuropathy commonly affects patients with chronic kidney disease, with
painful sensations in the feet, followed by numbness and weakness in the
legs and hands. The symptoms usually resolve following kidney
transplantation, but the mechanisms of uremic neuropathy and associated pain
symptoms remain unknown. As blood urea levels are elevated in patients with
chronic kidney disease, we examined the morphological and functional effects
of clinically observed levels of urea on sensory neurons. Methods Rat dorsal root ganglion neurons were treated with 10 or 50 mmol/L urea for
48 h, fixed and immunostained for PGP9.5 and βIII tubulin
immunofluorescence. Neurons were also immunostained for TRPV1, TRPM8 and
Gap43 expression, and the capsaicin sensitivity of urea- or vehicle-treated
neurons was determined. Results Urea-treated neurons had degenerating neurites with diminished PGP9.5
immunofluorescence, and swollen, retracted growth cones. βIII tubulin
appeared clumped after urea treatment. After 48 hours urea treatment,
neurite lengths were significantly reduced to 60 ± 2.6% (10 mmol/L,
**P < 0.01), and to 56.2 ± 3.3% (50 mmol/L, **P < 0.01), compared with
control neurons. Fewer neurons survived urea treatment, with 70.08 ± 13.3%
remaining after 10 mmol/L (*P < 0.05) and 61.49 ± 7.4% after 50 mmol/L
urea treatment (**P < 0.01), compared with controls. The proportion of
neurons expressing TRPV1 was reduced after urea treatment, but not TRPM8
expressing neurons. In functional studies, treatment with urea resulted in
dose-dependent neuronal sensitization. Capsaicin responses were
significantly increased to 115.29 ± 3.4% (10 mmol/L, **P < 0.01) and
125.3 ± 4.2% (50 mmol/L, **P < 0.01), compared with controls.
Sensitization due to urea was eliminated in the presence of the TRPV1
inhibitor SB705498, the mitogen-activated protein kinase kinase inhibitor
PD98059, the PI3 kinase inhibitor LY294002 and the TRPM8 inhibitor
N-(3-Aminopropyl)-2-[(3-methylphenyl)methoxy]-N-(2-thienylmethyl)benzamide
(AMTB hydrochloride). Conclusion Neurite degeneration and sensitization are consistent with uremic neuropathy
and provide a disease-relevant model to test new treatments.
Collapse
Affiliation(s)
- U Anand
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK.,Nanomedicine Research Laboratory, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Y Korchev
- Nanomedicine Research Laboratory, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - P Anand
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
73
|
Park SJ, Lee J, Lee S, Lim S, Noh J, Cho SY, Ha J, Kim H, Kim C, Park S, Lee DY, Kim E. Exposure of ultrafine particulate matter causes glutathione redox imbalance in the hippocampus: A neurometabolic susceptibility to Alzheimer's pathology. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 718:137267. [PMID: 32088476 DOI: 10.1016/j.scitotenv.2020.137267] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 06/10/2023]
Abstract
Particulate matter (PM) exposure is related to an increased risk of sporadic Alzheimer's disease (AD), the pathogenesis of which is explained by chronic neurometabolic disturbance. Therefore, PM-induced alterations in neurometabolism might herald AD. We aimed to identify brain region-specific changes in metabolic pathways associated with ultrafine particle (UFP) exposure and to determine whether such metabolic alterations are linked to susceptibility to AD. We constructed UFP exposure chambers and generated UFP by the pyrolysis method, which produces no toxic oxidized by-products of combustion, such as NOx and CO. Twenty male C57BL6 mice (11-12 months old) were exposed either to UFP or room air in the chambers for 3 weeks. One week following completion of UFP exposure, regional brain tissues, including the olfactory bulb, cortex, hippocampus, and cerebellum, were obtained and analyzed by metabolomics based on GC-MS and LC-MS, western blot analysis, and immunohistochemistry. Our results demonstrated that the metabolomic phenotype was distinct within the 4 different anatomical regions following UFP exposure. The highest level of metabolic change was identified in the hippocampus, a vulnerable region involved in AD pathogenesis. In this region, one of the key changes was perturbed redox homeostasis via alterations in the methionine-glutathione pathway. UFP exposure also induced oxidative stress and neuroinflammation, and importantly, increased Alzheimer's beta-amyloid levels in the hippocampus. These results suggest that inhaled UFP-induced perturbation in hippocampal redox homeostasis has a role in the pathogenesis of AD. Therefore, chronic exposure to UFP should be regarded as a cumulative environmental risk factor for sporadic AD.
Collapse
Affiliation(s)
- Soo Jin Park
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jimin Lee
- Department of Psychiatry, Institute of Behavioral Science in Medicine, BK21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seunghoon Lee
- Department of Mechanical Engineering, Dankook University, Gyeonggi-do, Yongin, Republic of Korea
| | - Sangchul Lim
- Department of Mechanical Engineering, Dankook University, Gyeonggi-do, Yongin, Republic of Korea
| | - Juhwan Noh
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Human Complexity and Systems Science, Yonsei University, Seoul, Republic of Korea
| | - So Yeon Cho
- Department of Psychiatry, Institute of Behavioral Science in Medicine, BK21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Junghee Ha
- Department of Psychiatry, Institute of Behavioral Science in Medicine, BK21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyunjeong Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, BK21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Changsoo Kim
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Institute of Human Complexity and Systems Science, Yonsei University, Seoul, Republic of Korea
| | - Sunho Park
- Department of Mechanical Engineering, Dankook University, Gyeonggi-do, Yongin, Republic of Korea.
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea.
| | - Eosu Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, BK21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
74
|
Xu J, Patassini S, Begley P, Church S, Waldvogel HJ, Faull RLM, Unwin RD, Cooper GJS. Cerebral deficiency of vitamin B5 (d-pantothenic acid; pantothenate) as a potentially-reversible cause of neurodegeneration and dementia in sporadic Alzheimer's disease. Biochem Biophys Res Commun 2020; 527:676-681. [PMID: 32416962 DOI: 10.1016/j.bbrc.2020.05.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/03/2020] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of age-related neurodegeneration and dementia, and there are no available treatments with proven disease-modifying actions. It is therefore appropriate to study hitherto-unknown aspects of brain structure/function in AD to seek alternative disease-related mechanisms that might be targeted by new therapeutic interventions with disease-modifying actions. During hypothesis-generating metabolomic studies of brain, we identified apparent differences in levels of vitamin B5 between AD cases and controls. We therefore developed a method based on gas chromatography-mass spectrometry by which we quantitated vitamin B5 concentrations in seven brain regions from nine AD cases and nine controls. We found that widespread, severe cerebral deficiency of vitamin B5 occurs in AD. This deficiency was worse in those regions known to undergo severe damage, including the hippocampus, entorhinal cortex, and middle temporal gyrus. Vitamin B5 is the obligate precursor of CoA/acetyl-CoA (acetyl-coenzyme A), which plays myriad key roles in the metabolism of all organs, including the brain. In brain, acetyl-CoA is the obligate precursor of the neurotransmitter acetylcholine, and the complex fatty-acyl groups that mediate the essential insulator role of myelin, both processes being defective in AD; moreover, the large cerebral vitamin B5 concentrations co-localize almost entirely to white matter. Vitamin B5 is well tolerated when administered orally to humans and other mammals. We conclude that cerebral vitamin B5 deficiency may well cause neurodegeneration and dementia in AD, which might be preventable or even reversible in its early stages, by treatment with suitable oral doses of vitamin B5.
Collapse
Affiliation(s)
- Jingshu Xu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9WL, UK; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Manchester Academic Health Sciences Centre, Manchester, UK; School of Biological Sciences, Faculty of Science, University of Auckland, New Zealand
| | - Stefano Patassini
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9WL, UK; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Manchester Academic Health Sciences Centre, Manchester, UK; School of Biological Sciences, Faculty of Science, University of Auckland, New Zealand
| | - Paul Begley
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9WL, UK; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Manchester Academic Health Sciences Centre, Manchester, UK
| | - Stephanie Church
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9WL, UK; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Manchester Academic Health Sciences Centre, Manchester, UK
| | - Henry J Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard D Unwin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9WL, UK; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Manchester Academic Health Sciences Centre, Manchester, UK
| | - Garth J S Cooper
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9WL, UK; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Manchester Academic Health Sciences Centre, Manchester, UK; School of Biological Sciences, Faculty of Science, University of Auckland, New Zealand; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
75
|
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
|
76
|
AMPfret: synthetic nanosensor for cellular energy states. Biochem Soc Trans 2020; 48:103-111. [PMID: 32010945 DOI: 10.1042/bst20190347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/14/2019] [Accepted: 01/10/2020] [Indexed: 12/22/2022]
Abstract
Cellular energy is a cornerstone of metabolism and is crucial for human health and disease. Knowledge of the cellular energy states and the underlying regulatory mechanisms is therefore key to understanding cell physiology and to design therapeutic interventions. Cellular energy states are characterised by concentration ratios of adenylates, in particular ATP:ADP and ATP:AMP. We applied synthetic biology approaches to design, engineer and validate a genetically encoded nano-sensor for cellular energy state, AMPfret. It employs the naturally evolved energy sensing of eukaryotic cells provided by the AMP-activated protein kinase (AMPK). Our synthetic nano-sensor relies on fluorescence resonance energy transfer (FRET) to detect changes in ATP:ADP and ATP:AMP ratios both in vitro and in cells in vivo. Construction and iterative optimisation relied on ACEMBL, a parallelised DNA assembly and construct screening technology we developed, facilitated by a method we termed tandem recombineering (TR). Our approach allowed rapid testing of numerous permutations of the AMPfret sensor to identify the most sensitive construct, which we characterised and validated both in the test tube and within cells.
Collapse
|
77
|
Ashraf A, Jeandriens J, Parkes HG, So PW. Iron dyshomeostasis, lipid peroxidation and perturbed expression of cystine/glutamate antiporter in Alzheimer's disease: Evidence of ferroptosis. Redox Biol 2020; 32:101494. [PMID: 32199332 PMCID: PMC7083890 DOI: 10.1016/j.redox.2020.101494] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Iron dyshomeostasis is implicated in Alzheimer’s disease (AD) alongside β-amyloid and tau pathologies. Despite the recent discovery of ferroptosis, an iron-dependent form cell death, hitherto, in vivo evidence of ferroptosis in AD is lacking. The present study uniquely adopts an integrated multi-disciplinary approach, combining protein (Western blot) and elemental analysis (total reflection X-ray fluorescence) with metabolomics (1H nuclear magnetic resonance spectroscopy) to identify iron dyshomeostasis and ferroptosis, and possible novel interactions with metabolic dysfunction in age-matched male cognitively normal (CN) and AD post-mortem brain tissue (n = 7/group). Statistical analysis was used to compute differences between CN and AD, and to examine associations between proteins, elements and/or metabolites. Iron dyshomeostasis with elevated levels of ferritin, in the absence of increased elemental iron, was observed in AD. Moreover, AD was characterised by enhanced expression of the light-chain subunit of the cystine/glutamate transporter (xCT) and lipid peroxidation, reminiscent of ferroptosis, alongside an augmented excitatory glutamate to inhibitory GABA ratio. Protein, element and metabolite associations also greatly differed between CN and AD suggesting widespread metabolic dysregulation in AD. We demonstrate iron dyshomeostasis, upregulated xCT (impaired glutathione metabolism) and lipid peroxidation in AD, suggesting anti-ferroptotic therapies may be efficacious in AD.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jérôme Jeandriens
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Department of Human Biology and Toxicology, Faculty of Medicine, University of Mons, Place du Parc 20, Mons, Belgium
| | - Harold G Parkes
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
78
|
Barthold D, Joyce G, Diaz Brinton R, Wharton W, Kehoe PG, Zissimopoulos J. Association of combination statin and antihypertensive therapy with reduced Alzheimer's disease and related dementia risk. PLoS One 2020; 15:e0229541. [PMID: 32130251 PMCID: PMC7055882 DOI: 10.1371/journal.pone.0229541] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/08/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Hyperlipidemia and hypertension are modifiable risk factors for Alzheimer's disease and related dementias (ADRD). Approximately 25% of adults over age 65 use both antihypertensives (AHTs) and statins for these conditions. While a growing body of evidence found statins and AHTs are independently associated with lower ADRD risk, no evidence exists on simultaneous use for different drug class combinations and ADRD risk. Our primary objective was to compare ADRD risk associated with concurrent use of different combinations of statins and antihypertensives. METHODS In a retrospective cohort study (2007-2014), we analyzed 694,672 Medicare beneficiaries in the United States (2,017,786 person-years) who concurrently used both statins and AHTs. Using logistic regression adjusting for age, socioeconomic status and comorbidities, we quantified incident ADRD diagnosis associated with concurrent use of different statin molecules (atorvastatin, pravastatin, rosuvastatin, and simvastatin) and AHT drug classes (two renin-angiotensin system (RAS)-acting AHTs, angiotensin converting enzyme inhibitors (ACEIs) or angiotensin-II receptor blockers (ARBs), vs non-RAS-acting AHTs). FINDINGS Pravastatin or rosuvastatin combined with RAS-acting AHTs reduce risk of ADRD relative to any statin combined with non-RAS-acting AHTs: ACEI+pravastatin odds ratio (OR) = 0.942 (CI: 0.899-0.986, p = 0.011), ACEI+rosuvastatin OR = 0.841 (CI: 0.794-0.892, p<0.001), ARB+pravastatin OR = 0.794 (CI: 0.748-0.843, p<0.001), ARB+rosuvastatin OR = 0.818 (CI: 0.765-0.874, p<0.001). ARBs combined with atorvastatin and simvastatin are associated with smaller reductions in risk, and ACEI with no risk reduction, compared to when combined with pravastatin or rosuvastatin. Among Hispanics, no combination of statins and RAS-acting AHTs reduces risk relative to combinations of statins and non-RAS-acting AHTs. Among blacks using ACEI+rosuvastatin, ADRD odds were 33% lower compared to blacks using other statins combined with non-RAS-acting AHTs (OR = 0.672 (CI: 0.548-0.825, p<0.001)). CONCLUSION Among older Americans, use of pravastatin and rosuvastatin to treat hyperlipidemia is less common than use of simvastatin and atorvastatin, however, in combination with RAS-acting AHTs, particularly ARBs, they may be more effective at reducing risk of ADRD. The number of Americans with ADRD may be reduced with drug treatments for vascular health that also confer effects on ADRD.
Collapse
Affiliation(s)
- Douglas Barthold
- Department of Pharmacy, The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington, Seattle, WA, United States of America
- * E-mail:
| | - Geoffrey Joyce
- School of Pharmacy, Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, CA, United States of America
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tuscon, AZ, United States of America
| | - Whitney Wharton
- School of Nursing, Emory University, Atlanta, GA, United States of America
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Patrick Gavin Kehoe
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Julie Zissimopoulos
- Price School of Public Policy, Schaeffer Center for Health Policy and Economics, University of Southern California, Los Angeles, CA, United States of America
| |
Collapse
|
79
|
Illiano P, Brambilla R, Parolini C. The mutual interplay of gut microbiota, diet and human disease. FEBS J 2020; 287:833-855. [DOI: 10.1111/febs.15217] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/21/2019] [Accepted: 01/16/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Placido Illiano
- The Miami Project to Cure Paralysis Department of Neurological Surgery University of Miami Miller School of Medicine FL USA
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis Department of Neurological Surgery University of Miami Miller School of Medicine FL USA
- Department of Neurobiology Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
- Department of Clinical Research BRIDGE‐Brain Research‐Inter‐Disciplinary Guided Excellence University of Southern Denmark Odense C Denmark
| | - Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Italy
| |
Collapse
|
80
|
APOE alters glucose flux through central carbon pathways in astrocytes. Neurobiol Dis 2020; 136:104742. [PMID: 31931141 DOI: 10.1016/j.nbd.2020.104742] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/04/2020] [Accepted: 01/08/2020] [Indexed: 02/02/2023] Open
Abstract
The Apolipoprotein E (APOE) gene is a major genetic risk factor associated with Alzheimer's disease (AD). APOE encodes for three main isoforms in humans (E2, E3, and E4). Homozygous E4 individuals have more than a 10-fold higher risk for developing late-onset AD, while E2 carriers are protected. A hallmark of AD is a reduction in cerebral glucose metabolism, alluding to a strong metabolic component in disease onset and progression. Interestingly, E4 individuals display a similar regional pattern of cerebral glucose hypometabolism decades prior to disease onset. Mapping this metabolic landscape may help elucidate the underlying biological mechanism of APOE-associated risk for AD. Efficient metabolic coupling of neurons and glia is necessary for proper neuronal function, and disruption in glial energy distribution has been proposed to contribute to neuronal cell death and AD pathology. One important function of astrocytes - canonically the primary source of apolipoprotein E in the brain - is to provide metabolic substrates (lactate, lipids, amino acids and neurotransmitters) to neurons. Here we investigate the effects of APOE on astrocyte glucose metabolism in vitro utilizing scintillation proximity assays, stable isotope tracer metabolomics, and gene expression analyses. Glucose uptake is impaired in E4 astrocytes relative to E2 or E3 with specific alterations in central carbon metabolism. Using stable isotope labeled glucose [U-13C] allowed analyses of astrocyte-specific deep metabolic networks affected by APOE, and provided insight to the effects downstream of glucose uptake. Enrichment of 13C in early steps of glycolysis was lowest in E4 astrocytes (highest in E2), while synthesis of lactate from glucose was highest in E4 astrocytes (lowest in E2). We observed an increase in glucose flux through the pentose phosphate pathway (PPP), with downstream increases in gluconeogenesis, lipid, and de novo nucleotide biosynthesis in E4 astrocytes. There was also a marked increase in 13C enrichment in the TCA cycle of E4 astrocytes - whose substrates were also incorporated into biosynthetic pathways at a higher rate. Pyruvate carboxylase (PC) and pyruvate dehydrogenase (PDH) are the two main enzymes controlling pyruvate entry to the TCA cycle. PC gene expression is increased in E4 astrocytes and the activity relative to PDH was also increased, compared to E2 or E3. Decreased enrichment in the TCA cycle of E2 and E3 astrocytes is suggestive of increased oxidation and non-glucose derived anaplerosis, which could be fueling mitochondrial ATP production. Conversely, E4 astrocytes appear to increase carbon flux into the TCA cycle to fuel cataplerosis. Together, these data demonstrate clear APOE isoform-specific effects on glucose utilization in astrocytes, including E4-associated increases in lactate synthesis, PPP flux, and de novo biosynthesis pathways.
Collapse
|
81
|
Dysregulation of multiple metabolic networks related to brain transmethylation and polyamine pathways in Alzheimer disease: A targeted metabolomic and transcriptomic study. PLoS Med 2020; 17:e1003012. [PMID: 31978055 PMCID: PMC6980402 DOI: 10.1371/journal.pmed.1003012] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 12/20/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND There is growing evidence that Alzheimer disease (AD) is a pervasive metabolic disorder with dysregulation in multiple biochemical pathways underlying its pathogenesis. Understanding how perturbations in metabolism are related to AD is critical to identifying novel targets for disease-modifying therapies. In this study, we test whether AD pathogenesis is associated with dysregulation in brain transmethylation and polyamine pathways. METHODS AND FINDINGS We first performed targeted and quantitative metabolomics assays using capillary electrophoresis-mass spectrometry (CE-MS) on brain samples from three groups in the Baltimore Longitudinal Study of Aging (BLSA) (AD: n = 17; Asymptomatic AD [ASY]: n = 13; Control [CN]: n = 13) (overall 37.2% female; mean age at death 86.118 ± 9.842 years) in regions both vulnerable and resistant to AD pathology. Using linear mixed-effects models within two primary brain regions (inferior temporal gyrus [ITG] and middle frontal gyrus [MFG]), we tested associations between brain tissue concentrations of 26 metabolites and the following primary outcomes: group differences, Consortium to Establish a Registry for Alzheimer's Disease (CERAD) (neuritic plaque burden), and Braak (neurofibrillary pathology) scores. We found significant alterations in concentrations of metabolites in AD relative to CN samples, as well as associations with severity of both CERAD and Braak, mainly in the ITG. These metabolites represented biochemical reactions in the (1) methionine cycle (choline: lower in AD, p = 0.003; S-adenosyl methionine: higher in AD, p = 0.005); (2) transsulfuration and glutathione synthesis (cysteine: higher in AD, p < 0.001; reduced glutathione [GSH]: higher in AD, p < 0.001); (3) polyamine synthesis/catabolism (spermidine: higher in AD, p = 0.004); (4) urea cycle (N-acetyl glutamate: lower in AD, p < 0.001); (5) glutamate-aspartate metabolism (N-acetyl aspartate: lower in AD, p = 0.002); and (6) neurotransmitter metabolism (gamma-amino-butyric acid: lower in AD, p < 0.001). Utilizing three Gene Expression Omnibus (GEO) datasets, we then examined mRNA expression levels of 71 genes encoding enzymes regulating key reactions within these pathways in the entorhinal cortex (ERC; AD: n = 25; CN: n = 52) and hippocampus (AD: n = 29; CN: n = 56). Complementing our metabolomics results, our transcriptomics analyses also revealed significant alterations in gene expression levels of key enzymatic regulators of biochemical reactions linked to transmethylation and polyamine metabolism. Our study has limitations: our metabolomics assays measured only a small proportion of all metabolites participating in the pathways we examined. Our study is also cross-sectional, limiting our ability to directly test how AD progression may impact changes in metabolite concentrations or differential-gene expression. Additionally, the relatively small number of brain tissue samples may have limited our power to detect alterations in all pathway-specific metabolites and their genetic regulators. CONCLUSIONS In this study, we observed broad dysregulation of transmethylation and polyamine synthesis/catabolism, including abnormalities in neurotransmitter signaling, urea cycle, aspartate-glutamate metabolism, and glutathione synthesis. Our results implicate alterations in cellular methylation potential and increased flux in the transmethylation pathways, increased demand on antioxidant defense mechanisms, perturbations in intermediate metabolism in the urea cycle and aspartate-glutamate pathways disrupting mitochondrial bioenergetics, increased polyamine biosynthesis and breakdown, as well as abnormalities in neurotransmitter metabolism that are related to AD.
Collapse
|
82
|
Ismail N, Kureishy N, Church SJ, Scholefield M, Unwin RD, Xu J, Patassini S, Cooper GJS. Vitamin B5 (d-pantothenic acid) localizes in myelinated structures of the rat brain: Potential role for cerebral vitamin B5 stores in local myelin homeostasis. Biochem Biophys Res Commun 2019; 522:220-225. [PMID: 31759626 PMCID: PMC6977085 DOI: 10.1016/j.bbrc.2019.11.052] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 01/15/2023]
Abstract
Vitamin B5 (d-pantothenic acid; pantothenate) is an essential trace nutrient that functions as the obligate precursor of coenzyme A (CoA), through which it plays key roles in myriad biological processes, including many that regulate carbohydrate, lipid, protein, and nucleic acid metabolism. In the brain, acetyl-CoA is necessary for synthesis of the complex fatty-acyl chains of myelin, and of the neurotransmitter acetylcholine. We recently found that cerebral pantothenate is markedly lowered, averaging ∼55% of control values in cases of Huntington's disease (HD) including those who are pre-symptomatic, and that regions where pantothenate is lowered correspond to those which are more severely damaged. Here we sought to determine the previously unknown distribution of pantothenate in the normal-rat brain, and whether the diabetic rat might be useful as a model for altered cerebral pantothenate metabolism. We employed histological staining (Nissl) to identify brain structures; immunohistochemistry with anti-pantothenate antibodies to determine the distribution of pantothenate in caudate putamen and cerebellum; and gas-chromatography/mass-spectrometry to quantitate levels of pantothenate and other metabolites in normal- and diabetic-rat brain. Remarkably, cerebral pantothenate was almost entirely localized to myelin-containing structures in both experimental groups. Diabetes did not modify levels or disposition of cerebral pantothenate. These findings are consistent with physiological localization of pantothenate in myelinated white-matter structures, where it could serve to support myelin synthesis. Further investigation of cerebral pantothenate is warranted in neurodegenerative diseases such as HD and Alzheimer's disease, where myelin loss is a known characteristic of pathogenesis.
Collapse
Affiliation(s)
- Nashwah Ismail
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK
| | - Nina Kureishy
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK
| | - Melissa Scholefield
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK; Stoller Biomarker Discovery Centre and Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, CityLabs 1.0 (3rd Floor), Nelson Street, XM13 9NQ, UK
| | - Jingshu Xu
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK
| | - Stefano Patassini
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK
| | - Garth J S Cooper
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M19 9NT, UK; School of Biological Sciences, Faculty of Science, University of Auckland, Private Bag 92 019, Auckland, 1142, New Zealand.
| |
Collapse
|
83
|
Tian X, Xie B, Zou Z, Jiao Y, Lin LE, Chen CL, Hsu CC, Peng J, Yang Z. Multimodal Imaging of Amyloid Plaques: Fusion of the Single-Probe Mass Spectrometry Image and Fluorescence Microscopy Image. Anal Chem 2019; 91:12882-12889. [PMID: 31536324 PMCID: PMC6885010 DOI: 10.1021/acs.analchem.9b02792] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. The formation of amyloid plaques by aggregated amyloid beta (Aβ) peptides is a primary event in AD pathology. Understanding the metabolomic features and related pathways is critical for studying plaque-related pathological events (e.g., cell death and neuron dysfunction). Mass spectrometry imaging (MSI), due to its high sensitivity and ability to obtain the spatial distribution of metabolites, has been applied to AD studies. However, limited studies of metabolites in amyloid plaques have been performed due to the drawbacks of the commonly used techniques such as matrix-assisted laser desorption/ionization MSI. In the current study, we obtained high spatial resolution (∼17 μm) MS images of the AD mouse brain using the Single-probe, a microscale sampling and ionization device, coupled to a mass spectrometer under ambient conditions. The adjacent slices were used to obtain fluorescence microscopy images to locate amyloid plaques. The MS image and the fluorescence microscopy image were fused to spatially correlate histological protein hallmarks with metabolomic features. The fused images produced significantly improved spatial resolution (∼5 μm), allowing for the determination of fine structures in MS images and metabolomic biomarkers representing amyloid plaques.
Collapse
Affiliation(s)
- Xiang Tian
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Boer Xie
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Zhu Zou
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Yun Jiao
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Li-En Lin
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Chih-Lin Chen
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
84
|
Chen K, Baluya D, Tosun M, Li F, Maletic-Savatic M. Imaging Mass Spectrometry: A New Tool to Assess Molecular Underpinnings of Neurodegeneration. Metabolites 2019; 9:E135. [PMID: 31295847 PMCID: PMC6681116 DOI: 10.3390/metabo9070135] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are prevalent and devastating. While extensive research has been done over the past decades, we are still far from comprehensively understanding what causes neurodegeneration and how we can prevent it or reverse it. Recently, systems biology approaches have led to a holistic examination of the interactions between genome, metabolome, and the environment, in order to shed new light on neurodegenerative pathogenesis. One of the new technologies that has emerged to facilitate such studies is imaging mass spectrometry (IMS). With its ability to map a wide range of small molecules with high spatial resolution, coupled with the ability to quantify them at once, without the need for a priori labeling, IMS has taken center stage in current research efforts in elucidating the role of the metabolome in driving neurodegeneration. IMS has already proven to be effective in investigating the lipidome and the proteome of various neurodegenerative diseases, such as Alzheimer's, Parkinson's, Huntington's, multiple sclerosis, and amyotrophic lateral sclerosis. Here, we review the IMS platform for capturing biological snapshots of the metabolic state to shed more light on the molecular mechanisms of the diseased brain.
Collapse
Affiliation(s)
- Kevin Chen
- Department of Biosciences, Rice University, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Dodge Baluya
- Chemical Imaging Research Core at MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Mehmet Tosun
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Feng Li
- Center for Drug Discovery and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.
- Department of Neuroscience and Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
85
|
Wu ZE, Kruger MC, Cooper GJS, Poppitt SD, Fraser K. Tissue-Specific Sample Dilution: An Important Parameter to Optimise Prior to Untargeted LC-MS Metabolomics. Metabolites 2019; 9:metabo9070124. [PMID: 31252691 PMCID: PMC6680868 DOI: 10.3390/metabo9070124] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 12/27/2022] Open
Abstract
When developing a sample preparation protocol for LC–MS untargeted metabolomics of a new sample matrix unfamiliar to the laboratory, selection of a suitable injection concentration is rarely described. Here we developed a simple workflow to address this issue prior to untargeted LC–MS metabolomics using pig adipose tissue and liver tissue. Bi-phasic extraction was performed to enable simultaneous optimisation of parameters for analysis of both lipids and polar extracts. A series of diluted pooled samples were analysed by LC–MS and used to evaluate signal linearity. Suitable injected concentrations were determined based on both the number of reproducible features and linear features. With our laboratory settings, the optimum concentrations of tissue mass to reconstitution solvent of liver and adipose tissue lipid fractions were found to be 125 mg/mL and 7.81 mg/mL respectively, producing 2811 (ESI+) and 4326 (ESI−) linear features from liver, 698 (ESI+) and 498 (ESI−) linear features from adipose tissue. For analysis of the polar fraction of both tissues, 250 mg/mL was suitable, producing 403 (ESI+) and 235 (ESI−) linear features from liver, 114 (ESI+) and 108 (ESI−) linear features from adipose tissue. Incorrect reconstitution volumes resulted in either severe overloading or poor linearity in our lipid data, while too dilute polar fractions resulted in a low number of reproducible features (<50) compared to hundreds of reproducible features from the optimum concentration used. Our study highlights on multiple matrices and multiple extract and chromatography types, the critical importance of determining a suitable injected concentration prior to untargeted LC–MS metabolomics, with the described workflow applicable to any matrix and LC–MS system.
Collapse
Affiliation(s)
- Zhanxuan E Wu
- Food Nutrition & Health, Food and Bio-based Products, AgResearch Limited, Palmerston North 4442, New Zealand
- School of Food and Advanced Technology, Massey University, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1142, New Zealand
| | - Marlena C Kruger
- School of Health Sciences, Massey University, Palmerston North 4442, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
| | - Garth J S Cooper
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, UK
- Human Nutrition Unit, School of Biological Sciences and Department of Medicine, University of Auckland, Auckland 1010, New Zealand
| | - Sally D Poppitt
- High-Value Nutrition National Science Challenge, Auckland 1142, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
- Human Nutrition Unit, School of Biological Sciences and Department of Medicine, University of Auckland, Auckland 1010, New Zealand
| | - Karl Fraser
- Food Nutrition & Health, Food and Bio-based Products, AgResearch Limited, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland 1142, New Zealand.
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand.
| |
Collapse
|
86
|
Patassini S, Begley P, Xu J, Church SJ, Kureishy N, Reid SJ, Waldvogel HJ, Faull RLM, Snell RG, Unwin RD, Cooper GJS. Cerebral Vitamin B5 (D-Pantothenic Acid) Deficiency as a Potential Cause of Metabolic Perturbation and Neurodegeneration in Huntington's Disease. Metabolites 2019; 9:E113. [PMID: 31212603 PMCID: PMC6630497 DOI: 10.3390/metabo9060113] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded CAG repeat in exon 1 of the HTT gene. HD usually manifests in mid-life with loss of GABAergic projection neurons from the striatum accompanied by progressive atrophy of the putamen followed by other brain regions, but linkages between the genetics and neurodegeneration are not understood. We measured metabolic perturbations in HD-human brain in a case-control study, identifying pervasive lowering of vitamin B5, the obligatory precursor of coenzyme A (CoA) that is essential for normal intermediary metabolism. Cerebral pantothenate deficiency is a newly-identified metabolic defect in human HD that could potentially: (i) impair neuronal CoA biosynthesis; (ii) stimulate polyol-pathway activity; (iii) impair glycolysis and tricarboxylic acid cycle activity; and (iv) modify brain-urea metabolism. Pantothenate deficiency could lead to neurodegeneration/dementia in HD that might be preventable by treatment with vitamin B5.
Collapse
Affiliation(s)
- Stefano Patassini
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Owlstone Medical, Cambridge Science Park, Cambridge CB4 0GJ, UK.
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Jingshu Xu
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
- Manchester Cancer Research Centre Building, The University of Manchester, Manchester M20 4GJ, UK.
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Nina Kureishy
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Suzanne J Reid
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Russell G Snell
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
| | - Garth J S Cooper
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M19 9NT, UK.
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand.
- Manchester Cancer Research Centre Building, The University of Manchester, Manchester M20 4GJ, UK.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
87
|
Primary fatty amides in plasma associated with brain amyloid burden, hippocampal volume, and memory in the European Medical Information Framework for Alzheimer's Disease biomarker discovery cohort. Alzheimers Dement 2019; 15:817-827. [PMID: 31078433 PMCID: PMC6849698 DOI: 10.1016/j.jalz.2019.03.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/06/2019] [Accepted: 03/04/2019] [Indexed: 12/24/2022]
Abstract
Introduction: A critical and as-yet unmet need in Alzheimer’s disease (AD) is the discovery of peripheral small molecule biomarkers. Given that brain pathology precedes clinical symptom onset, we set out to test whether metabolites in blood associated with pathology as indexed by cerebrospinal fluid (CSF) AD biomarkers. Methods: This study analyzed 593 plasma samples selected from the European Medical Information Framework for Alzheimer’s Disease Multimodal Biomarker Discovery study, of individuals who were cognitively healthy (n = 242), had mild cognitive impairment (n = 236), or had AD-type dementia (n = 115). Logistic regressions were carried out between plasma metabolites (n = 883) and CSF markers, magnetic resonance imaging, cognition, and clinical diagnosis. Results: Eight metabolites were associated with amyloid b and one with t-tau in CSF, these were primary fatty acid amides (PFAMs), lipokines, and amino acids. From these, PFAMs, glutamate, and aspartate also associated with hippocampal volume and memory. Discussion: PFAMs have been found increased and associated with amyloid b burden in CSF and clinical measures.
Collapse
|
88
|
Hadar A, Gurwitz D. Peripheral transcriptomic biomarkers for early detection of sporadic Alzheimer disease? DIALOGUES IN CLINICAL NEUROSCIENCE 2019. [PMID: 30936769 PMCID: PMC6436957 DOI: 10.31887/dcns.2018.20.4/dgurwitz] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Alzheimer disease (AD) is the major epidemic of the 21st century, its prevalence rising along with improved human longevity. Early AD diagnosis is key to successful treatment, as currently available therapeutics only allow small benefits for diagnosed AD patients. By contrast, future therapeutics, including those already in preclinical or clinical trials, are expected to afford neuroprotection prior to widespread brain damage and dementia. Brain imaging technologies are developing as promising tools for early AD diagnostics, yet their high cost limits their utility for screening at-risk populations. Blood or plasma transcriptomics, proteomics, and/or metabolomics may pave the way for cost-effective AD risk screening in middle-aged individuals years ahead of cognitive decline. This notion is exemplified by data mining of blood transcriptomics from a published dataset. Consortia blood sample collection and analysis from large cohorts with mild cognitive impairment followed longitudinally for their cognitive state would allow the development of a reliable and inexpensive early AD screening tool.
Collapse
Affiliation(s)
- Adva Hadar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 69978 Israel
| |
Collapse
|
89
|
Wang T, Cheng Y, Han H, Liu J, Tian B, Liu X. miR-194 Accelerates Apoptosis of Aβ 1⁻42-Transduced Hippocampal Neurons by Inhibiting Nrn1 and Decreasing PI3K/Akt Signaling Pathway Activity. Genes (Basel) 2019; 10:genes10040313. [PMID: 31010100 PMCID: PMC6523401 DOI: 10.3390/genes10040313] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022] Open
Abstract
This article explores the mechanism of miR-194 on the proliferation and apoptosis of Aβ1–42-transduced hippocampal neurons. Aβ1–42-transduced hippocampal neuron model was established by inducing hippocampal neurons with Aβ1–42. MTT assay and flow cytometry were used to detect the viability and apoptosis of hippocampal neurons, respectively. qRT-PCR was used to detect changes in miR-194 and Nrn1 expression after Aβ1–42 induction. Aβ1–42-transduced hippocampal neurons were transfected with miR-194 mimics and/or Nrn1 overexpression vectors. Their viability and neurite length were detected by MTT assay and immunofluorescence, respectively. Western blot was used to detect protein expression. Aβ1–42 inhibited Aβ1–42-transduced hippocampal neuron activity and promoted their apoptosis in a dose-dependent manner. miR-194 was upregulated and Nrn1 was downregulated in Aβ1–42-transduced hippocampal neurons (p < 0.05). Compared with the model group, Aβ1–42-transduced hippocampal neurons of the miR-194 mimic group had much lower activity, average longest neurite length, Nrn1, p-AkT, and Bcl-2 protein expression and had much higher Bax, Caspase-3, and Cleaved Caspase-3 protein expression. Compared with the model group, Aβ1–42-transduced hippocampal neurons of the LV-Nrn1 group had much higher activity, average longest neurite length, Nrn1, p-AkT, and Bcl-2 protein expression and had much lower Bax, Caspase-3, and Cleaved Caspase-3 protein expression. Nrn1 is a target gene of miR-194. miR-194 inhibited apoptosis of Aβ1–42-transduced hippocampal neurons by inhibiting Nrn1 and decreasing PI3K/AkT signaling pathway activity.
Collapse
Affiliation(s)
- Tingting Wang
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Yaling Cheng
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Haibin Han
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Jie Liu
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Bo Tian
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Xiaocui Liu
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| |
Collapse
|
90
|
Krystofova J, Pathipati P, Russ J, Sheldon A, Ferriero D. The Arginase Pathway in Neonatal Brain Hypoxia-Ischemia. Dev Neurosci 2019; 40:437-450. [PMID: 30995639 PMCID: PMC6784534 DOI: 10.1159/000496467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Brain damage after hypoxia-ischemia (HI) occurs in an age-dependent manner. Neuroprotective strategies assumed to be effective in adults might have deleterious effects in the immature brain. In order to create effective therapies, the complex pathophysiology of HI in the developing brain requires exploring new mechanisms. Critical determinants of neuronal survival after HI are the extent of vascular dysfunction, inflammation, and oxidative stress, followed later by tissue repair. The key enzyme of these processes in the human body is arginase (ARG) that acts via the bioavailability of nitric oxide, and the synthesis of polyamines and proline. ARG is expressed throughout the brain in different cells. However, little is known about the effect of ARG in pathophysiological states of the brain, especially hypoxia-ischemia. Here, we summarize the role of ARG during neurodevelopment as well as in various brain pathologies.
Collapse
Affiliation(s)
- Jana Krystofova
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA,
| | - Praneeti Pathipati
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey Russ
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Ann Sheldon
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Donna Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
91
|
Sade D, Shaham-Niv S, Arnon ZA, Tavassoly O, Gazit E. Seeding of proteins into amyloid structures by metabolite assemblies may clarify certain unexplained epidemiological associations. Open Biol 2019; 8:rsob.170229. [PMID: 29367352 PMCID: PMC5795054 DOI: 10.1098/rsob.170229] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
The accumulation of various metabolites appears to be associated with diverse human diseases. However, the aetiological link between metabolic alteration and the observed diseases is still elusive. This includes the correlation between the abnormally high levels of homocysteine and quinolinic acid in Alzheimer's disease, as well as the accumulation of oncometabolites in malignant processes. Here, we suggest and discuss a possible mechanistic insight into metabolite accumulation in conditions such as neurodegenerative diseases and cancer. Our hypothesis is based on the demonstrated ability of metabolites to form amyloid-like structures in inborn error of metabolism disorders and the potential of such metabolite amyloids to promote protein aggregation. This notion can provide a new paradigm for neurodegeneration and cancer, as both conditions were linked to loss of function due to protein aggregation. Similar to the well-established observation of amyloid formation in many degenerative disorders, the formation of amyloids by tumour-suppressor proteins, including p53, was demonstrated in malignant states. Moreover, this new paradigm could fill the gap in understanding the high occurrence of specific types of cancer among genetic error of metabolism patients. This hypothesis offers a fresh view on the aetiology of some of the most abundant human maladies and may redirect the efforts towards new therapeutic developments.
Collapse
Affiliation(s)
- Dorin Sade
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shira Shaham-Niv
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Zohar A Arnon
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Omid Tavassoly
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel .,Sagol Interdisciplinary School of Neurosciences, Tel Aviv University, Tel Aviv 6997801, Israel.,Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
92
|
Xu J, Patassini S, Rustogi N, Riba-Garcia I, Hale BD, Phillips AM, Waldvogel H, Haines R, Bradbury P, Stevens A, Faull RLM, Dowsey AW, Cooper GJS, Unwin RD. Regional protein expression in human Alzheimer's brain correlates with disease severity. Commun Biol 2019; 2:43. [PMID: 30729181 PMCID: PMC6361956 DOI: 10.1038/s42003-018-0254-9] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 12/03/2018] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that currently affects 36 million people worldwide with no effective treatment available. Development of AD follows a distinctive pattern in the brain and is poorly modelled in animals. Therefore, it is vital to widen the spatial scope of the study of AD and prioritise the study of human brains. Here we show that functionally distinct human brain regions display varying and region-specific changes in protein expression. These changes provide insights into the progression of disease, novel AD-related pathways, the presence of a gradient of protein expression change from less to more affected regions and a possibly protective protein expression profile in the cerebellum. This spatial proteomics analysis provides a framework which can underpin current research and open new avenues to enhance molecular understanding of AD pathophysiology, provide new targets for intervention and broaden the conceptual frameworks for future AD research.
Collapse
Affiliation(s)
- Jingshu Xu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Private Bag 92019, Auckland, 1142 New Zealand
| | - Stefano Patassini
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Private Bag 92019, Auckland, 1142 New Zealand
| | - Nitin Rustogi
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| | - Isabel Riba-Garcia
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| | - Benjamin D. Hale
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| | - Alexander M Phillips
- Department of Electrical Engineering and Electronics, University of Liverpool, Liverpool, L69 3GJ UK
| | - Henry Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142 New Zealand
| | - Robert Haines
- Research IT, The University of Manchester, Manchester, M13 9PL UK
| | - Phil Bradbury
- Research IT, The University of Manchester, Manchester, M13 9PL UK
| | - Adam Stevens
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PL UK
| | - Richard L. M. Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142 New Zealand
| | - Andrew W. Dowsey
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- Department of Population Health Sciences and Bristol Veterinary School, Faculty of Health Sciences, University of Bristol, Bristol, BS8 2BN UK
| | - Garth J. S. Cooper
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
- School of Biological Sciences, and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Private Bag 92019, Auckland, 1142 New Zealand
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142 New Zealand
| | - Richard D. Unwin
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, Core Technology Facility (3rd Floor), 46 Grafton Street, Manchester, M13 9NT UK
| |
Collapse
|
93
|
Alpay Savasan Z, Yilmaz A, Ugur Z, Aydas B, Bahado-Singh RO, Graham SF. Metabolomic Profiling of Cerebral Palsy Brain Tissue Reveals Novel Central Biomarkers and Biochemical Pathways Associated with the Disease: A Pilot Study. Metabolites 2019; 9:metabo9020027. [PMID: 30717353 PMCID: PMC6409919 DOI: 10.3390/metabo9020027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/17/2022] Open
Abstract
Cerebral palsy (CP) is one of the most common causes of motor disability in childhood, with complex and heterogeneous etiopathophysiology and clinical presentation. Understanding the metabolic processes associated with the disease may aid in the discovery of preventive measures and therapy. Tissue samples (caudate nucleus) were obtained from post-mortem CP cases (n = 9) and age- and gender-matched control subjects (n = 11). We employed a targeted metabolomics approach using both 1H NMR and direct injection liquid chromatography-tandem mass spectrometry (DI/LC-MS/MS). We accurately identified and quantified 55 metabolites using 1H NMR and 186 using DI/LC-MS/MS. Among the 222 detected metabolites, 27 showed significant concentration changes between CP cases and controls. Glycerophospholipids and urea were the most commonly selected metabolites used to develop predictive models capable of discriminating between CP and controls. Metabolomics enrichment analysis identified folate, propanoate, and androgen/estrogen metabolism as the top three significantly perturbed pathways. We report for the first time the metabolomic profiling of post-mortem brain tissue from patients who died from cerebral palsy. These findings could help to further investigate the complex etiopathophysiology of CP while identifying predictive, central biomarkers of CP.
Collapse
Affiliation(s)
- Zeynep Alpay Savasan
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Division, Beaumont Health System, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
- Oakland University-William Beaumont School of Medicine, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Ali Yilmaz
- Beaumont Research Institute, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Zafer Ugur
- Beaumont Research Institute, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Buket Aydas
- Departments of Mathematics and Computer Sciences, Albion College, 611 E. Porter St., Albion, MI 49224, USA.
| | - Ray O Bahado-Singh
- Department of Obstetrics and Gynecology, Maternal Fetal Medicine Division, Beaumont Health System, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
- Oakland University-William Beaumont School of Medicine, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| | - Stewart F Graham
- Oakland University-William Beaumont School of Medicine, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
- Beaumont Research Institute, Beaumont Health, 3811 W. 13 Mile Road, Royal Oak, MI 48073, USA.
| |
Collapse
|
94
|
Polis B, Srikanth KD, Gurevich V, Gil-Henn H, Samson AO. L-Norvaline, a new therapeutic agent against Alzheimer's disease. Neural Regen Res 2019; 14:1562-1572. [PMID: 31089055 PMCID: PMC6557086 DOI: 10.4103/1673-5374.255980] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Growing evidence highlights the role of arginase activity in the manifestation of Alzheimer’s disease (AD). Upregulation of arginase was shown to contribute to neurodegeneration. Regulation of arginase activity appears to be a promising approach for interfering with the pathogenesis of AD. Therefore, the enzyme represents a novel therapeutic target. In this study, we administered an arginase inhibitor, L-norvaline (250 mg/L), for 2.5 months to a triple-transgenic model (3×Tg-AD) harboring PS1M146V, APPSwe, and tauP301L transgenes. Then, the neuroprotective effects of L-norvaline were evaluated using immunohistochemistry, proteomics, and quantitative polymerase chain reaction assays. Finally, we identified the biological pathways activated by the treatment. Remarkably, L-norvaline treatment reverses the cognitive decline in AD mice. The treatment is neuroprotective as indicated by reduced beta-amyloidosis, alleviated microgliosis, and reduced tumor necrosis factor transcription levels. Moreover, elevated levels of neuroplasticity related postsynaptic density protein 95 were detected in the hippocampi of mice treated with L-norvaline. Furthermore, we disclosed several biological pathways, which were involved in cell survival and neuroplasticity and were activated by the treatment. Through these modes of action, L-norvaline has the potential to improve the symptoms of AD and even interferes with its pathogenesis. As such, L-norvaline is a promising neuroprotective molecule that might be tailored for the treatment of a range of neurodegenerative disorders. The study was approved by the Bar-Ilan University Animal Care and Use Committee (approval No. 82-10-2017) on October 1, 2017.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine; Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Kolluru D Srikanth
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Vyacheslav Gurevich
- Laboratory of Cancer Personalized Medicine and Diagnostic Genomics, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Abraham O Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
95
|
Hadar A. Peripheral transcriptomic biomarkers for early detection of sporadic Alzheimer disease? DIALOGUES IN CLINICAL NEUROSCIENCE 2018; 20:293-300. [PMID: 30936769 PMCID: PMC6436957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Alzheimer disease (AD) is the major epidemic of the 21st century, its prevalence rising along with improved human longevity. Early AD diagnosis is key to successful treatment, as currently available therapeutics only allow small benefits for diagnosed AD patients. By contrast, future therapeutics, including those already in preclinical or clinical trials, are expected to afford neuroprotection prior to widespread brain damage and dementia. Brain imaging technologies are developing as promising tools for early AD diagnostics, yet their high cost limits their utility for screening at-risk populations. Blood or plasma transcriptomics, proteomics, and/or metabolomics may pave the way for cost-effective AD risk screening in middle-aged individuals years ahead of cognitive decline. This notion is exemplified by data mining of blood transcriptomics from a published dataset. Consortia blood sample collection and analysis from large cohorts with mild cognitive impairment followed longitudinally for their cognitive state would allow the development of a reliable and inexpensive early AD screening tool.
Collapse
Affiliation(s)
- Adva Hadar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine
| |
Collapse
|
96
|
Paraskevaidi M, Morais CLM, Halliwell DE, Mann DMA, Allsop D, Martin-Hirsch PL, Martin FL. Raman Spectroscopy to Diagnose Alzheimer's Disease and Dementia with Lewy Bodies in Blood. ACS Chem Neurosci 2018; 9:2786-2794. [PMID: 29865787 DOI: 10.1021/acschemneuro.8b00198] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Accurate identification of Alzheimer's disease (AD) is still of major clinical importance considering the current lack of noninvasive and low-cost diagnostic approaches. Detection of early stage AD is particularly desirable as it would allow early intervention or recruitment of patients into clinical trials. There is also an unmet need for discrimination of AD from dementia with Lewy bodies (DLB), as many cases of the latter are misdiagnosed as AD. Biomarkers based on a simple blood test would be useful in research and clinical practice. Raman spectroscopy has been implemented to analyze blood plasma of a cohort that consisted of early stage AD, late-stage AD, DLB, and healthy controls. Classification algorithms achieved high accuracy for the different groups: early stage AD vs healthy with 84% sensitivity, 86% specificity; late-stage AD vs healthy with 84% sensitivity, 77% specificity; DLB vs healthy with 83% sensitivity, 87% specificity; early-stage AD vs DLB with 81% sensitivity, 88% specificity; late-stage AD vs DLB with 90% sensitivity, 93% specificity; and lastly, early-stage AD vs late-stage AD 66% sensitivity and 83% specificity. G-score values were also estimated between 74% and 91%, demonstrating that the overall performance of the classification model was satisfactory. The wavenumbers responsible for differentiation were assigned to important biomolecules, which can serve as a panel of biomarkers. These results suggest a cost-effective, blood-based test for neurodegeneration in dementias.
Collapse
Affiliation(s)
- Maria Paraskevaidi
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, U.K
| | - Camilo L. M. Morais
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, U.K
| | - Diane E. Halliwell
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, U.K
| | - David M. A. Mann
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, University of Manchester, Greater Manchester Neurosciences Centre, Salford Royal Hospital, Salford M6 8HD, U.K
| | - David Allsop
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, U.K
| | - Pierre L. Martin-Hirsch
- Department of Obstetrics and Gynaecology, Central Lancashire Teaching Hospitals NHS Foundation Trust, Preston PR2 9HT, U.K
| | - Francis L. Martin
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, U.K
| |
Collapse
|
97
|
Possible Clues for Brain Energy Translation via Endolysosomal Trafficking of APP-CTFs in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2764831. [PMID: 30420907 PMCID: PMC6215552 DOI: 10.1155/2018/2764831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/14/2018] [Accepted: 08/19/2018] [Indexed: 02/07/2023]
Abstract
Vascular dysfunctions, hypometabolism, and insulin resistance are high and early risk factors for Alzheimer's disease (AD), a leading neurological disease associated with memory decline and cognitive dysfunctions. Early defects in glucose transporters and glycolysis occur during the course of AD progression. Hypometabolism begins well before the onset of early AD symptoms; this timing implicates the vulnerability of hypometabolic brain regions to beta-secretase 1 (BACE-1) upregulation, oxidative stress, inflammation, synaptic failure, and cell death. Despite the fact that ketone bodies, astrocyte-neuron lactate shuttle, pentose phosphate pathway (PPP), and glycogenolysis compensate to provide energy to the starving AD brain, a considerable energy crisis still persists and increases during disease progression. Studies that track brain energy metabolism in humans, animal models of AD, and in vitro studies reveal striking upregulation of beta-amyloid precursor protein (β-APP) and carboxy-terminal fragments (CTFs). Currently, the precise role of CTFs is unclear, but evidence supports increased endosomal-lysosomal trafficking of β-APP and CTFs through autophagy through a vague mechanism. While intracellular accumulation of Aβ is attributed as both the cause and consequence of a defective endolysosomal-autophagic system, much remains to be explored about the other β-APP cleavage products. Many recent works report altered amino acid catabolism and expression of several urea cycle enzymes in AD brains, but the precise cause for this dysregulation is not fully explained. In this paper, we try to connect the role of CTFs in the energy translation process in AD brain based on recent findings.
Collapse
|
98
|
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]
|
99
|
Muguruma Y, Tsutsui H, Noda T, Akatsu H, Inoue K. Widely targeted metabolomics of Alzheimer's disease postmortem cerebrospinal fluid based on 9-fluorenylmethyl chloroformate derivatized ultra-high performance liquid chromatography tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1091:53-66. [PMID: 29852382 DOI: 10.1016/j.jchromb.2018.05.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/10/2018] [Accepted: 05/21/2018] [Indexed: 12/22/2022]
Abstract
Confirmed biomarkers of postmortem cerebrospinal fluid (pCSF) are used to differentiate between Alzheimer's disease (AD) patients and healthy seniors with high diagnostic accuracy. However, the extent to which the performance of specific metabolic profiling facilitates reliable estimations of the concentrations of the different pCSF biomarkers and their ratios remains unclear. The interpretation of the lower levels of molecules of metabolic profiling and their concentration ratios in pCSF related to brain disorders could facilitate an unchallenging detection of peripheral biomarkers of AD stages and other dementia types. In this study, we proposed the use of widely targeted metabolomics for pCSF metabolic profiling using 9-fluorenylmethyl chloroformate- (FMOC) derivatized ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) to evaluate the diversity of 97 amine-mediated metabolic patterns and pathways from confirmed diagnosis based on AD brain pathology. Our results identified the metabolites that contributed toward and mutually influenced the principal component analysis plot with integrated analytes. Furthermore, the AD group showed a significant variation in several analyte concentration levels compared to those of control subjects. These trends of the concentration levels expressed by the amine metabolic pathways indicated the decreased activity of polyamine and tryptophan-kynurenine (Trp-Kyn) metabolisms. Moreover, increased metabolites such as methionine sulfoxide, 3-methoxy-anthranilate, cadaverine, guanine, and histamine were observed by widely targeted metabolomics of pCSF from the AD subjects. According to their metabolic pathway analysis using FMOC-derivatized UHPLC-MS/MS assay, we supposed that the involvement of polyamine and Trp-Kyn metabolisms was observed in the pCSF samples.
Collapse
Affiliation(s)
- Yoshio Muguruma
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan
| | - Haruhito Tsutsui
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan; ONO Pharmaceutical Co., Ltd, 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Takumi Noda
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan; ONO Pharmaceutical Co., Ltd, 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Hiroyasu Akatsu
- Department of Medicine for Aging Place, Community Health Care/Community-Based Medical Education, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-0001, Japan; Department of Neuropathology, Choju Medical Institute, Fukushimura Hospital, Toyohashi 441-8124, Japan
| | - Koichi Inoue
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga 525-8577, Japan.
| |
Collapse
|
100
|
Cabrera D, Kruger M, Wolber FM, Roy NC, Totman JJ, Henry CJ, Cameron-Smith D, Fraser K. Association of Plasma Lipids and Polar Metabolites with Low Bone Mineral Density in Singaporean-Chinese Menopausal Women: A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1045. [PMID: 29789485 PMCID: PMC5982084 DOI: 10.3390/ijerph15051045] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/09/2018] [Accepted: 05/19/2018] [Indexed: 01/23/2023]
Abstract
The diagnosis of osteoporosis is mainly based on clinical examination and bone mineral density assessments. The present pilot study compares the plasma lipid and polar metabolite profiles in blood plasma of 95 Singaporean-Chinese (SC) menopausal women with normal and low bone mineral density (BMD) using an untargeted metabolomic approach. The primary finding of this study was the association between lipids and femoral neck BMD in SC menopausal women. Twelve lipids were identified to be associated with low BMD by the orthogonal partial least squares (OPLS) model. Plasma concentrations of eight glycerophospholipid, glycerolipid, and sphingolipid species were significantly lower in menopausal women with low BMD but higher in two glycerophospholipid species (phosphatidylinositol and phosphatidic acid). Further, this study found no significant differences in plasma amino acid metabolites. However, trends for lower 4-aminobutyric acid, turanose, proline, aminopropionitrile, threonine, and methionine were found in women with low BMD. This pilot study identified associations between lipid metabolism and femoral neck BMD in SC women. Further studies are required on larger populations for evaluating the bone health effect of these compounds and their usefulness as clinical biomarkers for osteoporosis prediction in women.
Collapse
Affiliation(s)
- Diana Cabrera
- School of Food and Nutrition, Massey University, Tennent Drive, Palmerston North 4442, New Zealand.
- Food Nutrition & Health Team, Food & Bio-Based Products Group, AgResearch Grasslands, Palmerston North 4442, New Zealand.
| | - Marlena Kruger
- School of Food and Nutrition, Massey University, Tennent Drive, Palmerston North 4442, New Zealand.
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand.
| | - Frances M Wolber
- Centre for Metabolic Health Research, Massey University, Tennent Drive, Palmerston North 4442, New Zealand.
| | - Nicole C Roy
- Food Nutrition & Health Team, Food & Bio-Based Products Group, AgResearch Grasslands, Palmerston North 4442, New Zealand.
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland 1142, New Zealand.
| | - John J Totman
- A*Star-NUS Clinical Imaging Research Centre, Singapore 117599, Singapore.
| | | | - David Cameron-Smith
- Food Nutrition & Health Team, Food & Bio-Based Products Group, AgResearch Grasslands, Palmerston North 4442, New Zealand.
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand.
- The Liggins Institute, The University of Auckland, Auckland 1142, New Zealand.
| | - Karl Fraser
- Food Nutrition & Health Team, Food & Bio-Based Products Group, AgResearch Grasslands, Palmerston North 4442, New Zealand.
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland 1142, New Zealand.
| |
Collapse
|