1
|
Andersen JV. The Glutamate/GABA-Glutamine Cycle: Insights, Updates, and Advances. J Neurochem 2025; 169:e70029. [PMID: 40066661 PMCID: PMC11894596 DOI: 10.1111/jnc.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Synaptic homeostasis of the principal neurotransmitters glutamate and GABA is tightly regulated by an intricate metabolic coupling between neurons and astrocytes known as the glutamate/GABA-glutamine cycle. In this cycle, astrocytes take up glutamate and GABA from the synapse and convert these neurotransmitters into glutamine. Astrocytic glutamine is subsequently transferred to neurons, serving as the principal precursor for neuronal glutamate and GABA synthesis. The glutamate/GABA-glutamine cycle integrates multiple cellular processes, including neurotransmitter release, uptake, synthesis, and metabolism. All of these processes are deeply interdependent and closely coupled to cellular energy metabolism. Astrocytes display highly active mitochondrial oxidative metabolism and several unique metabolic features, including glycogen storage and pyruvate carboxylation, which are essential to sustain continuous glutamine release. However, new roles of oligodendrocytes and microglia in neurotransmitter recycling are emerging. Malfunction of the glutamate/GABA-glutamine cycle can lead to severe synaptic disruptions and may be implicated in several brain diseases. Here, I review central aspects and recent advances of the glutamate/GABA-glutamine cycle to highlight how the cycle is functionally connected to critical brain functions and metabolism. First, an overview of glutamate, GABA, and glutamine transport is provided in relation to neurotransmitter recycling. Then, central metabolic aspects of the glutamate/GABA-glutamine cycle are reviewed, with a special emphasis on the critical metabolic roles of glial cells. Finally, I discuss how aberrant neurotransmitter recycling is linked to neurodegeneration and disease, focusing on astrocyte metabolic dysfunction and brain lipid homeostasis as emerging pathological mechanisms. Instead of viewing the glutamate/GABA-glutamine cycle as individual biochemical processes, a more holistic and integrative approach is needed to advance our understanding of how neurotransmitter recycling modulates brain function in both health and disease.
Collapse
Affiliation(s)
- Jens V. Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
2
|
Al-Thani NA, Zinck D, Stewart GS, Costello DA. Modulation of Urea Transport Attenuates TLR2-Mediated Microglial Activation and Upregulates Microglial Metabolism In Vitro. Metabolites 2024; 14:634. [PMID: 39590870 PMCID: PMC11596256 DOI: 10.3390/metabo14110634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Alzheimer's disease (AD) is a neurodegenerative disorder traditionally characterised by the presence of amyloid beta (Aβ) plaques and neurofibrillary tau tangles in the brain. However, emerging research has highlighted additional metabolic hallmarks of AD pathology. These include the metabolic reprogramming of microglia in favour of glycolysis over oxidative phosphorylation. This shift is attributed to an 'M1'-like pro-inflammatory phenotype, which exacerbates neuroinflammation and contributes to neuronal damage. The urea cycle also presents as an altered metabolic pathway in AD, due to elevated urea levels and altered expression of urea cycle enzymes, metabolites, and transporters in the brain. However, to date, these changes remain largely unexplored. Methods: This study focuses on understanding the effects of extracellular urea and urea transporter-B (UT-B) inhibition on inflammatory changes in lipoteichoic acid (LTA)-stimulated BV2 microglia and on the viability of SH-SY5Y neuronal cells under oxidative stress and neurotoxic conditions. Results: In BV2 microglia, UT-B inhibition demonstrated a notable anti-inflammatory effect by reducing the formation of nitric oxide (NO) and the expression of tumour necrosis factor α (TNFα) and CCL2 in response to stimulation with the toll-like receptor (TLR)2 agonist, lipoteichoic acid (LTA). This was accompanied by a reduction in extracellular urea and upregulation of UT-B expression. The application of exogenous urea was also shown to mediate the inflammatory profile of BV2 cells in a similar manner but had only a modest impact on UT-B expression. While exposure to LTA alone did not alter the microglial metabolic profile, inhibition of UT-B upregulated the expression of genes associated with both glycolysis and fatty acid oxidation. Conversely, neither increased extracellular urea nor UT-B inhibition had a significant impact on cell viability or cytotoxicity in SH-SY5Y neurones exposed to oxidative stressors tert-butyl hydroperoxide (t-BHP) and 6-hydroxydopamine (6-OHDA). Conclusions: This study further highlights the involvement of urea transport in regulating the neuroinflammation associated with AD. Moreover, we reveal a novel role for UT-B in maintaining microglial metabolic homeostasis. Taken together, these findings contribute supporting evidence to the regulation of UT-B as a therapeutic target for intervention into neuroinflammatory and neurodegenerative disease.
Collapse
Affiliation(s)
- Najlaa A. Al-Thani
- School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland; (N.A.A.-T.); (D.Z.)
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Dylan Zinck
- School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland; (N.A.A.-T.); (D.Z.)
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Gavin S. Stewart
- School of Biology and Environmental Science, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Derek A. Costello
- School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland; (N.A.A.-T.); (D.Z.)
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
3
|
Zouridis S, Nasir AB, Aspichueta P, Syn WK. The Link between Metabolic Syndrome and the Brain. Digestion 2024:1-9. [PMID: 39369701 DOI: 10.1159/000541696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Metabolic syndrome (MetS) is a cluster of cardiometabolic conditions that has been linked to high risk for cardiovascular disease, liver complications, and several malignancies. More recently, MetS has been associated with cognitive dysfunction. SUMMARY Studies have shown an association with minimal cognitive impairment, progression to vascular dementia, and even Alzheimer's disease. MetS components have been individually explored, and glucose intolerance has the strongest association with impairment in several cognitive domains. Several hypotheses have been proposed regarding the pathophysiology underlying the MetS-cognitive dysfunction association, and even though insulin resistance plays a major role, more studies are needed to elucidate this topic. Moreover, several other factors contributing to this association have been identified. Liver disease and more specifically metabolic dysfunction-associated steatotic liver disease can on its own contribute to cognitive decline through systemic inflammation and higher ammonia levels. Gut dysbiosis that has also been identified in MetS can also lead to cognitive impairment through several mechanisms that result in neurotoxicity. Finally, there are several other factors that may modify the MetS-cognitive dysfunction relationship, such as lifestyle, diet, education status, and age. More recently, circadian syndrome was explored and was found to be even more strongly associated with cognitive impairment. KEY MESSAGE MetS is associated with cognitive decline. Certain cardiometabolic risk factors have a stronger association with cognitive impairment, and there are several factors that may modify this relationship. The aim of this review was to assess and summarize the existing body of evidence on the association between MetS and cognitive impairment and identify areas that necessitate further investigation.
Collapse
Affiliation(s)
- Spyridon Zouridis
- Division of Gastroenterology and Hepatology, Saint Louis University, St. Louis, Missouri, USA,
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain,
| | - Ahmad Basil Nasir
- Division of Gastroenterology and Hepatology, Saint Louis University, St. Louis, Missouri, USA
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Wing-Kin Syn
- Division of Gastroenterology and Hepatology, Saint Louis University, St. Louis, Missouri, USA
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| |
Collapse
|
4
|
Lyon AC, Lippa CF, Eiser AR. Metabolic and Environmental Biomarkers in Mild Cognitive Impairment and Dementia: An Exploratory Study. JOURNAL OF INTEGRATIVE AND COMPLEMENTARY MEDICINE 2024; 30:793-801. [PMID: 38330435 DOI: 10.1089/jicm.2023.0583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Objective: To determine the frequency with which suspected pathogenic factors, including metals and metabolites that might contribute to Alzheimer's disease (AD), may be found in patients with cognitive impairment through commonly available blood tests. Methods: A variety of serum studies, including metals, ammonia, homocysteine, vitamin B12, folate, thyroid tests, metabolic products, and inflammatory markers, were measured in two cohorts: one meeting mild cognitive impairment (MCI) criteria and the other meeting mild-to-moderate dementia (DE) criteria. Medications these patients received were reviewed. Results: Metal abnormalities were detected in over half the subjects, including evidence of mercury, lead, and arsenic elevation as well as instances of excessive essential metals, iron (Fe), and copper. Some metal aberration was detected in 64% of the DE group and 66% of the MCI group. Females were more likely to have elevated copper, consistent with hormonal effects on copper excretion. Homocysteinemia was the most common abnormality, detected in 71% with DE and 67% with MCI, while methylmalonic acid was not elevated. Slight hyperammonemia was moderately common (38%) suggesting a hepatic factor in this subset. Findings of moderate insulin resistance were present in nearly half (44% DE, 52% MCI). Sixty of 65 (92%) had at least one abnormal biomarker and 60% had two or more. The most common drug taken by the total cohort was proton pump inhibitors at 22% DE and 38% MCI. Conclusions: This study suggests that both toxic metals and excessive vital metals such as copper and iron, as well as common metabolic and hepatic factors are detectable at both stages of MCI and DE. There appears to be a multiplicity of provocative factors leading to DE. Individualized interventions based on these parameters may be a means to reduce cognitive decline leading to DE. A more comprehensive prospective study of these environmental and metabolic factors with corrective early interventions appears warranted.
Collapse
Affiliation(s)
- Abigail C Lyon
- Department of Neurology, Cognitive Disorders & Comprehensive Alzheimer's Disease Center, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Carol F Lippa
- Department of Neurology, Cognitive Disorders & Comprehensive Alzheimer's Disease Center, Vickie & Jack Farber Institute for Neuroscience, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Arnold R Eiser
- Senior Scholar, Penn Center for Public Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Lachén-Montes M, Cartas-Cejudo P, Cortés A, Anaya-Cubero E, Peral E, Ausín K, Díaz-Peña R, Fernández-Irigoyen J, Santamaría E. Involvement of Glucosamine 6 Phosphate Isomerase 2 (GNPDA2) Overproduction in β-Amyloid- and Tau P301L-Driven Pathomechanisms. Biomolecules 2024; 14:394. [PMID: 38672412 PMCID: PMC11048700 DOI: 10.3390/biom14040394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative olfactory disorder affecting millions of people worldwide. Alterations in the hexosamine- or glucose-related pathways have been described through AD progression. Specifically, an alteration in glucosamine 6 phosphate isomerase 2 (GNPDA2) protein levels has been observed in olfactory areas of AD subjects. However, the biological role of GNPDA2 in neurodegeneration remains unknown. Using mass spectrometry, multiple GNPDA2 interactors were identified in human nasal epithelial cells (NECs) mainly involved in intraciliary transport. Moreover, GNPDA2 overexpression induced an increment in NEC proliferation rates, accompanied by transcriptomic alterations in Type II interferon signaling or cellular stress responses. In contrast, the presence of beta-amyloid or mutated Tau-P301L in GNPDA2-overexpressing NECs induced a slowdown in the proliferative capacity in parallel with a disruption in protein processing. The proteomic characterization of Tau-P301L transgenic zebrafish embryos demonstrated that GNPDA2 overexpression interfered with collagen biosynthesis and RNA/protein processing, without inducing additional changes in axonal outgrowth defects or neuronal cell death. In humans, a significant increase in serum GNPDA2 levels was observed across multiple neurological proteinopathies (AD, Lewy body dementia, progressive supranuclear palsy, mixed dementia and amyotrophic lateral sclerosis) (n = 215). These data shed new light on GNPDA2-dependent mechanisms associated with the neurodegenerative process beyond the hexosamine route.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario Universitario de Navarra (HUN), IdiSNA, Navarra Institute for Health Research, Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain; (M.L.-M.); (P.C.-C.); (A.C.); (E.A.-C.); (E.P.); (K.A.); (R.D.-P.); (J.F.-I.)
| |
Collapse
|
6
|
Andersson OP. Is the microbiome the cause of irritable bowel syndrome and inflammatory bowel disease? Lessons to consider from odontology. Int J Colorectal Dis 2023; 38:117. [PMID: 37150763 DOI: 10.1007/s00384-023-04406-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/10/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND A substantial amount of research is pointing to the disrupted microbiome and dysfunctional host-microbiome interaction as potential causes of Irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). The true cause of the diseases is still not fully elucidated, and the various treatments used are not truly effective in the long run, especially for IBD, since a true cure is not known to exist. Treatment failure and surgery are common for IBD, many times leading to a perceived lower quality of life, not to mention the enormous cost for society for treatment up until that point and after. Although it is clear that the microbiome has a major role in the disease, it seems the majority of the research and treatments are still focused on treating and understanding the inflammation and not the primary cause of the inflammation in the first place. This was also the case for many decades in the search for the cause of periodontitis (PD) and gingivitis (GV), a destructive and non-destructive inflammatory disorder, respectively, the first resulting in loss of tissue supporting the teeth. There was much uncertainty and confusion until it was fully established that the microbiome was the cause. PD treatments primarily nowadays reflect the cause, i.e. the removal of microbes. There is no doubt, however, that the inflammatory pathways are important in both diseases and the purpose of this text is not to dispute this in respect to gastrointestinal disorders too. However, a different view on inflammation and associated disorders is explored to explain the nature of extraintestinal manifestations. PURPOSE The aim of this report is not to systematically fully review the literature to try to strengthen causality, as there are many reviews that explore the microbial aspects of IBS and IBD. Instead, the objective is to above all reflect on what has been learned in the field of odontology/stomatology and discuss relevant gastrointestinal research in order to propose tentative hypotheses and questions regarding IBS and IBD aetiology. Perhaps it could help soften the confusion regarding the microbial aetiology and dysbiosis concept, while guiding future research and treatments, primarily regarding microbial transplants, antibiotics, and diet.
Collapse
|
7
|
Sharma A, Kumar R, Varadwaj P. Smelling the Disease: Diagnostic Potential of Breath Analysis. Mol Diagn Ther 2023; 27:321-347. [PMID: 36729362 PMCID: PMC9893210 DOI: 10.1007/s40291-023-00640-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2023] [Indexed: 02/03/2023]
Abstract
Breath analysis is a relatively recent field of research with much promise in scientific and clinical studies. Breath contains endogenously produced volatile organic components (VOCs) resulting from metabolites of ingested precursors, gut and air-passage bacteria, environmental contacts, etc. Numerous recent studies have suggested changes in breath composition during the course of many diseases, and breath analysis may lead to the diagnosis of such diseases. Therefore, it is important to identify the disease-specific variations in the concentration of breath to diagnose the diseases. In this review, we explore methods that are used to detect VOCs in laboratory settings, VOC constituents in exhaled air and other body fluids (e.g., sweat, saliva, skin, urine, blood, fecal matter, vaginal secretions, etc.), VOC identification in various diseases, and recently developed electronic (E)-nose-based sensors to detect VOCs. Identifying such VOCs and applying them as disease-specific biomarkers to obtain accurate, reproducible, and fast disease diagnosis could serve as an alternative to traditional invasive diagnosis methods. However, the success of VOC-based identification of diseases is limited to laboratory settings. Large-scale clinical data are warranted for establishing the robustness of disease diagnosis. Also, to identify specific VOCs associated with illness states, extensive clinical trials must be performed using both analytical instruments and electronic noses equipped with stable and precise sensors.
Collapse
Affiliation(s)
- Anju Sharma
- Systems Biology Lab, Indian Institute of Information Technology, Allahabad, Uttar Pradesh, India
| | - Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Uttar Pradesh, Lucknow Campus, Lucknow, India
| | - Pritish Varadwaj
- Systems Biology Lab, Indian Institute of Information Technology, Allahabad, Uttar Pradesh, India.
| |
Collapse
|
8
|
Strefeler A, Jan M, Quadroni M, Teav T, Rosenberg N, Chatton JY, Guex N, Gallart-Ayala H, Ivanisevic J. Molecular insights into sex-specific metabolic alterations in Alzheimer's mouse brain using multi-omics approach. Alzheimers Res Ther 2023; 15:8. [PMID: 36624525 PMCID: PMC9827669 DOI: 10.1186/s13195-023-01162-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by altered cellular metabolism in the brain. Several of these alterations have been found to be exacerbated in females, known to be disproportionately affected by AD. We aimed to unravel metabolic alterations in AD at the metabolic pathway level and evaluate whether they are sex-specific through integrative metabolomic, lipidomic, and proteomic analysis of mouse brain tissue. METHODS We analyzed male and female triple-transgenic mouse whole brain tissue by untargeted mass spectrometry-based methods to obtain a molecular signature consisting of polar metabolite, complex lipid, and protein data. These data were analyzed using multi-omics factor analysis. Pathway-level alterations were identified through joint pathway enrichment analysis or by separately evaluating lipid ontology and known proteins related to lipid metabolism. RESULTS Our analysis revealed significant AD-associated and in part sex-specific alterations across the molecular signature. Sex-dependent alterations were identified in GABA synthesis, arginine biosynthesis, and in alanine, aspartate, and glutamate metabolism. AD-associated alterations involving lipids were also found in the fatty acid elongation pathway and lysophospholipid metabolism, with a significant sex-specific effect for the latter. CONCLUSIONS Through multi-omics analysis, we report AD-associated and sex-specific metabolic alterations in the AD brain involving lysophospholipid and amino acid metabolism. These findings contribute to the characterization of the AD phenotype at the molecular level while considering the effect of sex, an overlooked yet determinant metabolic variable.
Collapse
Affiliation(s)
- Abigail Strefeler
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Maxime Jan
- grid.9851.50000 0001 2165 4204Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Manfredo Quadroni
- grid.9851.50000 0001 2165 4204Protein Analysis Facility, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Tony Teav
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Nadia Rosenberg
- grid.9851.50000 0001 2165 4204Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jean-Yves Chatton
- grid.9851.50000 0001 2165 4204Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Guex
- grid.9851.50000 0001 2165 4204Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Hector Gallart-Ayala
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| |
Collapse
|
9
|
Neuffer J, González-Domínguez R, Lefèvre-Arbogast S, Low DY, Driollet B, Helmer C, Du Preez A, de Lucia C, Ruigrok SR, Altendorfer B, Aigner L, Lucassen PJ, Korosi A, Thuret S, Manach C, Pallàs M, Urpi-Sardà M, Sánchez-Pla A, Andres-Lacueva C, Samieri C. Exploration of the Gut-Brain Axis through Metabolomics Identifies Serum Propionic Acid Associated with Higher Cognitive Decline in Older Persons. Nutrients 2022; 14:4688. [PMID: 36364950 PMCID: PMC9655149 DOI: 10.3390/nu14214688] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
The gut microbiome is involved in nutrient metabolism and produces metabolites that, via the gut−brain axis, signal to the brain and influence cognition. Human studies have so far had limited success in identifying early metabolic alterations linked to cognitive aging, likely due to limitations in metabolite coverage or follow-ups. Older persons from the Three-City population-based cohort who had not been diagnosed with dementia at the time of blood sampling were included, and repeated measures of cognition over 12 subsequent years were collected. Using a targeted metabolomics platform, we identified 72 circulating gut-derived metabolites in a case−control study on cognitive decline, nested within the cohort (discovery n = 418; validation n = 420). Higher serum levels of propionic acid, a short-chain fatty acid, were associated with increased odds of cognitive decline (OR for 1 SD = 1.40 (95% CI 1.11, 1.75) for discovery and 1.26 (1.02, 1.55) for validation). Additional analyses suggested mediation by hypercholesterolemia and diabetes. Propionic acid strongly correlated with blood glucose (r = 0.79) and with intakes of meat and cheese (r > 0.15), but not fiber (r = 0.04), suggesting a minor role of prebiotic foods per se, but a possible link to processed foods, in which propionic acid is a common preservative. The adverse impact of propionic acid on metabolism and cognition deserves further investigation.
Collapse
Affiliation(s)
- Jeanne Neuffer
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Raúl González-Domínguez
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sophie Lefèvre-Arbogast
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Dorrain Y. Low
- Human Nutrition Unit, Université Clermont Auvergne, INRAEUMR1019, F-63000 Clermont Ferrand, France
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Bénédicte Driollet
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Catherine Helmer
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| | - Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Chiara de Lucia
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
| | - Silvie R. Ruigrok
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Paul J. Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
- The Center for Urban Mental Health, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9NU, UK
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Claudine Manach
- Human Nutrition Unit, Université Clermont Auvergne, INRAEUMR1019, F-63000 Clermont Ferrand, France
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, 08028 Barcelona, Spain
| | - Mireia Urpi-Sardà
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alex Sánchez-Pla
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Andres-Lacueva
- Nutrition, Food Science and Gastronomy Department, Food Innovation Network (XIA), Institute of Nutrition and Food Safety (INSA-UB), Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cécilia Samieri
- Bordeaux Population Health Research Center, University of Bordeaux, INSERMUMR 1219, F-33000 Bordeaux, France
| |
Collapse
|
10
|
Amphiphysin AoRvs167-Mediated Membrane Curvature Facilitates Trap Formation, Endocytosis, and Stress Resistance in Arthrobotrysoligospora. Pathogens 2022; 11:pathogens11090997. [PMID: 36145429 PMCID: PMC9501185 DOI: 10.3390/pathogens11090997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Bin1/Amphiphysin/Rvs (BAR) domain-containing proteins mediate fundamental cellular processes, including membrane remodeling and endocytosis. Nematode-trapping (NT) fungi can differentiate to form trapping structures through highly reorganized cell membranes and walls. In this study, we identified the NT fungus Arthrobotrys oligospora ortholog of yeast Rvs167 and documented its involvement in membrane bending and endocytosis. We further confirmed that the deletion of AoRvs167 makes the fungus more hypersensitive to osmotic salt (Nacl), higher temperatures (28 to 30 °C), and the cell wall perturbation agent Congo red. In addition, the disruption of AoRvs167 reduced the trap formation capacity. Hence, AoRvs167 may regulate fungal pathogenicity through the integrity of plasma membranes and cell walls.
Collapse
|
11
|
Andersen JV, Schousboe A, Verkhratsky A. Astrocyte energy and neurotransmitter metabolism in Alzheimer's disease: integration of the glutamate/GABA-glutamine cycle. Prog Neurobiol 2022; 217:102331. [PMID: 35872221 DOI: 10.1016/j.pneurobio.2022.102331] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023]
Abstract
Astrocytes contribute to the complex cellular pathology of Alzheimer's disease (AD). Neurons and astrocytes function in close collaboration through neurotransmitter recycling, collectively known as the glutamate/GABA-glutamine cycle, which is essential to sustain neurotransmission. Neurotransmitter recycling is intimately linked to astrocyte energy metabolism. In the course of AD, astrocytes undergo extensive metabolic remodeling, which may profoundly affect the glutamate/GABA-glutamine cycle. The consequences of altered astrocyte function and metabolism in relation to neurotransmitter recycling are yet to be comprehended. Metabolic alterations of astrocytes in AD deprive neurons of metabolic support, thereby contributing to synaptic dysfunction and neurodegeneration. In addition, several astrocyte-specific components of the glutamate/GABA-glutamine cycle, including glutamine synthesis and synaptic neurotransmitter uptake, are perturbed in AD. Integration of the complex astrocyte biology within the context of AD is essential for understanding the fundamental mechanisms of the disease, while restoring astrocyte metabolism may serve as an approach to arrest or even revert clinical progression of AD.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania.
| |
Collapse
|
12
|
Ammonia induces amyloidogenesis in astrocytes by promoting amyloid precursor protein translocation into the endoplasmic reticulum. J Biol Chem 2022; 298:101933. [PMID: 35427648 PMCID: PMC9117890 DOI: 10.1016/j.jbc.2022.101933] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 03/26/2022] [Accepted: 03/26/2022] [Indexed: 11/25/2022] Open
Abstract
Hyperammonemia is known to cause various neurological dysfunctions such as seizures and cognitive impairment. Several studies have suggested that hyperammonemia may also be linked to the development of Alzheimer’s disease (AD). However, the direct evidence for a role of ammonia in the pathophysiology of AD remains to be discovered. Herein, we report that hyperammonemia increases the amount of mature amyloid precursor protein (mAPP) in astrocytes, the largest and most prevalent type of glial cells in the central nervous system that are capable of metabolizing glutamate and ammonia, and promotes amyloid beta (Aβ) production. We demonstrate the accumulation of mAPP in astrocytes was primarily due to enhanced endocytosis of mAPP from the plasma membrane. A large proportion of internalized mAPP was targeted not to the lysosome, but to the endoplasmic reticulum, where processing enzymes β-secretase BACE1 (beta-site APP cleaving enzyme 1) and γ-secretase presenilin-1 are expressed, and mAPP is cleaved to produce Aβ. Finally, we show the ammonia-induced production of Aβ in astrocytic endoplasmic reticulum was specific to Aβ42, a principal component of senile plaques in AD patients. Our studies uncover a novel mechanism of Aβ42 production in astrocytes and also provide the first evidence that ammonia induces the pathogenesis of AD by regulating astrocyte function.
Collapse
|
13
|
Eldridge RC, Uppal K, Shokouhi M, Smith MR, Hu X, Qin ZS, Jones DP, Hajjar I. Multiomics Analysis of Structural Magnetic Resonance Imaging of the Brain and Cerebrospinal Fluid Metabolomics in Cognitively Normal and Impaired Adults. Front Aging Neurosci 2022; 13:796067. [PMID: 35145393 PMCID: PMC8822333 DOI: 10.3389/fnagi.2021.796067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/27/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Integrating brain imaging with large scale omics data may identify novel mechanisms of mild cognitive impairment (MCI) and early Alzheimer's disease (AD). We integrated and analyzed brain magnetic resonance imaging (MRI) with cerebrospinal fluid (CSF) metabolomics to elucidate metabolic mechanisms and create a "metabolic map" of the brain in prodromal AD. METHODS In 145 subjects (85 cognitively normal controls and 60 with MCI), we derived voxel-wise gray matter volume via whole-brain structural MRI and conducted high-resolution untargeted metabolomics on CSF. Using a data-driven approach consisting of partial least squares discriminant analysis, a multiomics network clustering algorithm, and metabolic pathway analysis, we described dysregulated metabolic pathways in CSF mapped to brain regions associated with MCI in our cohort. RESULTS The multiomics network algorithm clustered metabolites with contiguous imaging voxels into seven distinct communities corresponding to the following brain regions: hippocampus/parahippocampal gyrus (three distinct clusters), thalamus, posterior thalamus, parietal cortex, and occipital lobe. Metabolic pathway analysis indicated dysregulated metabolic activity in the urea cycle, and many amino acids (arginine, histidine, lysine, glycine, tryptophan, methionine, valine, glutamate, beta-alanine, and purine) was significantly associated with those regions (P < 0.05). CONCLUSION By integrating CSF metabolomics data with structural MRI data, we linked specific AD-susceptible brain regions to disrupted metabolic pathways involving nitrogen excretion and amino acid metabolism critical for cognitive function. Our findings and analytical approach may extend drug and biomarker research toward more multiomics approaches.
Collapse
Affiliation(s)
- Ronald C. Eldridge
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Mahsa Shokouhi
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, United States
| | - M. Ryan Smith
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Xin Hu
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Zhaohui S. Qin
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Dean P. Jones
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Ihab Hajjar
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
14
|
Salcedo C, Andersen JV, Vinten KT, Pinborg LH, Waagepetersen HS, Freude KK, Aldana BI. Functional Metabolic Mapping Reveals Highly Active Branched-Chain Amino Acid Metabolism in Human Astrocytes, Which Is Impaired in iPSC-Derived Astrocytes in Alzheimer's Disease. Front Aging Neurosci 2021; 13:736580. [PMID: 34603012 PMCID: PMC8484639 DOI: 10.3389/fnagi.2021.736580] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/04/2021] [Indexed: 01/04/2023] Open
Abstract
The branched-chain amino acids (BCAAs) leucine, isoleucine, and valine are important nitrogen donors for synthesis of glutamate, the main excitatory neurotransmitter in the brain. The glutamate carbon skeleton originates from the tricarboxylic acid (TCA) cycle intermediate α-ketoglutarate, while the amino group is derived from nitrogen donors such as the BCAAs. Disturbances in neurotransmitter homeostasis, mainly of glutamate, are strongly implicated in the pathophysiology of Alzheimer's disease (AD). The divergent BCAA metabolism in different cell types of the human brain is poorly understood, and so is the involvement of astrocytic and neuronal BCAA metabolism in AD. The goal of this study is to provide the first functional characterization of BCAA metabolism in human brain tissue and to investigate BCAA metabolism in AD pathophysiology using astrocytes and neurons derived from human-induced pluripotent stem cells (hiPSCs). Mapping of BCAA metabolism was performed using mass spectrometry and enriched [15N] and [13C] isotopes of leucine, isoleucine, and valine in acutely isolated slices of surgically resected cerebral cortical tissue from human brain and in hiPSC-derived brain cells carrying mutations in either amyloid precursor protein (APP) or presenilin-1 (PSEN-1). We revealed that both human astrocytes of acutely isolated cerebral cortical slices and hiPSC-derived astrocytes were capable of oxidatively metabolizing the carbon skeleton of BCAAs, particularly to support glutamine synthesis. Interestingly, hiPSC-derived astrocytes with APP and PSEN-1 mutations exhibited decreased amino acid synthesis of glutamate, glutamine, and aspartate derived from leucine metabolism. These results clearly demonstrate that there is an active BCAA metabolism in human astrocytes, and that leucine metabolism is selectively impaired in astrocytes derived from the hiPSC models of AD. This impairment in astrocytic BCAA metabolism may contribute to neurotransmitter and energetic imbalances in the AD brain.
Collapse
Affiliation(s)
- Claudia Salcedo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Tore Vinten
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars H Pinborg
- Epilepsy Clinic and Neurobiology Research Unit, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer's disease? Anal Biochem 2021; 636:114387. [PMID: 34537182 DOI: 10.1016/j.ab.2021.114387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.
Collapse
|
16
|
Cheon SY, Song J. The Association between Hepatic Encephalopathy and Diabetic Encephalopathy: The Brain-Liver Axis. Int J Mol Sci 2021; 22:ijms22010463. [PMID: 33466498 PMCID: PMC7796499 DOI: 10.3390/ijms22010463] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatic encephalopathy (HE) is one of the main consequences of liver disease and is observed in severe liver failure and cirrhosis. Recent studies have provided significant evidence that HE shows several neurological symptoms including depressive mood, cognitive dysfunction, impaired circadian rhythm, and attention deficits as well as motor disturbance. Liver disease is also a risk factor for the development of diabetes mellitus. Diabetic encephalopathy (DE) is characterized by cognitive dysfunction and motor impairment. Recent research investigated the relationship between metabolic changes and the pathogenesis of neurological disease, indicating the importance between metabolic organs and the brain. Given that a diverse number of metabolites and changes in the brain contribute to neurologic dysfunction, HE and DE are emerging types of neurologic disease. Here, we review significant evidence of the association between HE and DE, and summarise the common risk factors. This review may provide promising therapeutic information and help to design a future metabolic organ-related study in relation to HE and DE.
Collapse
Affiliation(s)
- So Yeong Cheon
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Korea;
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Korea
- Correspondence: ; Tel.: +82-61-379-2706; Fax: +82-61-375-5834
| |
Collapse
|
17
|
The nuclear factor kappa B (NF-κB) signaling pathway is involved in ammonia-induced mitochondrial dysfunction. Mitochondrion 2020; 57:63-75. [PMID: 33378713 DOI: 10.1016/j.mito.2020.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022]
Abstract
Hyperammonemia is very toxic to the brain, leading to inflammation, disruption of brain cellular energy metabolism and cognitive function. However, the underlying mechanism(s) for these impairments is still not fully understood. This study investigated the effects of ammonia in hippocampal astroglia derived from C57BL/6 mice. Parameters measured included oxygen consumption rates (OCR), ATP, cytochrome c oxidase (COX) activity, alterations in oxidative phosphorylation (OXPHOS), nuclear factor kappa B (NF-κB) subunits, key regulators of mitochondrial biogenesis (peroxisome proliferator-activated receptor gamma coactivator1-alpha (PGC-1α), calcium/calmodulin-dependent protein kinase II (CaMKII), cAMP-response element binding protein (CREB), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), early growth response (Egr) factor family of proteins, and mitochondrial transcription factor A (TFAM). Ammonia was found to decrease mitochondrial numbers, potentially through a CaMKII-CREB-PGC1α-Nrf2 pathway in astroglia. Ammonia did not alter the levels of Egrs and TFAM in astroglia. Ammonia decreased OCR, ATP, COX, and OXPHOS levels in astroglia. To assess whether energy metabolism is reduced by ammonia through NF-κB associated pathways, astroglia were treated with ammonia alone or with NF-κB inhibitors such as Bay11-7082 or SN50. Mitochondrial OCR levels were reduced in the presence of NF-κB inhibitors; however co-treatment of NF-κB inhibitors and ammonia reversed mitochondrial deficits. Further, ammonia increased translocation of the NF-κB p65 into the nucleus of astroglia that correlates with an increased activity of NF-κB. These findings suggest that the NF-κB signaling pathway is putatively involved in ammonia-induced changes in bioenergetics in astroglia. Such research has critical implications for the treatment of disorders in which brain bioenergetics is compromised.
Collapse
|
18
|
Mitrayana, Nikita JG, Wasono MAJ, Satriawan M. CO2 laser photoacoustic spectrometer for measuring ethylene, acetone, and ammonia in the breath of patients with renal disease. SENSING AND BIO-SENSING RESEARCH 2020. [DOI: 10.1016/j.sbsr.2020.100387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
19
|
Andersen JV, Christensen SK, Westi EW, Diaz-delCastillo M, Tanila H, Schousboe A, Aldana BI, Waagepetersen HS. Deficient astrocyte metabolism impairs glutamine synthesis and neurotransmitter homeostasis in a mouse model of Alzheimer's disease. Neurobiol Dis 2020; 148:105198. [PMID: 33242587 DOI: 10.1016/j.nbd.2020.105198] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) leads to cerebral accumulation of insoluble amyloid-β plaques causing synaptic dysfunction and neuronal death. Neurons rely on astrocyte-derived glutamine for replenishment of the amino acid neurotransmitter pools. Perturbations of astrocyte glutamine synthesis have been described in AD, but whether this functionally affects neuronal neurotransmitter synthesis is not known. Since the synthesis and recycling of neurotransmitter glutamate and GABA are intimately coupled to cellular metabolism, the aim of this study was to provide a functional investigation of neuronal and astrocytic energy and neurotransmitter metabolism in AD. To achieve this, we incubated acutely isolated cerebral cortical and hippocampal slices from 8-month-old female 5xFAD mice, in the presence of 13C isotopically enriched substrates, with subsequent gas chromatography-mass spectrometry (GC-MS) analysis. A prominent neuronal hypometabolism of [U-13C]glucose was observed in the hippocampal slices of the 5xFAD mice. Investigating astrocyte metabolism, using [1,2-13C]acetate, revealed a marked reduction in glutamine synthesis, which directly hampered neuronal synthesis of GABA. This was supported by an increased metabolism of exogenously supplied [U-13C]glutamine, suggesting a neuronal metabolic compensation of the reduced astrocytic glutamine supply. In contrast, astrocytic metabolism of [U-13C]GABA was reduced, whereas [U-13C]glutamate metabolism was unaffected. Finally, astrocyte de novo synthesis of glutamate and glutamine was hampered, whereas the enzymatic capacity of glutamine synthetase for ammonia fixation was maintained. Collectively, we demonstrate that deficient astrocyte metabolism leads to reduced glutamine synthesis, directly impairing neuronal GABA synthesis in the 5xFAD brain. These findings suggest that astrocyte metabolic dysfunction may be fundamental for the imbalances of synaptic excitation and inhibition in the AD brain.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Sofie K Christensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Marta Diaz-delCastillo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Heikki Tanila
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
20
|
Grishin D, Kasap E, Izotov A, Lisitsa A. Multifaceted ammonia transporters. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1812443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- D.V. Grishin
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - E.Y. Kasap
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - A.A. Izotov
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - A.V. Lisitsa
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| |
Collapse
|
21
|
Instant activation of TRP channels by NH 4 + promotes neuronal bursting and glutamate spikes in CA1 neurons. Curr Res Physiol 2020; 3:20-29. [PMID: 34746817 PMCID: PMC8562246 DOI: 10.1016/j.crphys.2020.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/11/2020] [Accepted: 05/24/2020] [Indexed: 11/22/2022] Open
Abstract
Ammonia ( NH 4 + ) is a by-product of cell metabolism and may elicit subcellular effects with specific physiological responses. Chronic effects have been implicated in several neurological diseases and attributed to persistent elevation in blood ammonia levels transferred to the brain. In previous studies the activities of neurons and astrocytes have been examined at ammonia concentrations an order of magnitude higher than measured in the blood. The effects developed within several minutes. Here we focused upon acute responses of neurons to ammonia and whether they may occur at much lower doses. To this end, we combined patch-clamp in CA1 neurons with glutamate imaging in hippocampal slices. Particular attention was paid to the Rett syndrome that has been originally attributed to hyperammonemia. We compared the responses in the wild-type (WT) and model Rett mice (MECP2-null, RTT) to ammonia doses from 0.3 mM on. In both preparations NH 4 + promptly depolarized neurons and increased the ambient glutamate. The bursting activity emerged in WT and it was augmented in RTT. Searching for subcellular mechanisms we examined possible modulation of ion channels by ammonia. We did not find any changes in HCN- and Ca2+ currents, which substantially contribute to the bursting activity. The non-selective cation channels were markedly potentiated by ammonia. ASIC channels had a major contribution to the augmentation of neuronal activity by ammonia. Interestingly, their general blocker amiloride (100 μM) moderately excited CA1 cells akin to NH 4 + . In its presence subsequent ammonia effects were markedly compromised. Blockade of TRPC1 channels partially occluded NH 4 + effects. ASIC and TRPC1 blockers decreased the amplitude of excitatory postsynaptic currents (EPSC) and neuronal bursts, congruent with a postsynaptic location of the channels. Inhibition of TRPV1 channels potentiated the responses to NH 4 + . EPSC amplitudes did not change, but the frequency decreased, indicating presynaptic effects. All extracellular NH 4 + actions were observed at concentrations as low as 0.3 mM and the neurons reacted immediately after ammonia arrived the slice. We propose that a brief augmentation of neuronal activity by NH 4 + may occur either spontaneously during arousal or induced by inhalation of smelling salts.
Collapse
|
22
|
Griffin JWD, Bradshaw PC. Effects of a high protein diet and liver disease in an in silico model of human ammonia metabolism. Theor Biol Med Model 2019; 16:11. [PMID: 31366360 PMCID: PMC6670211 DOI: 10.1186/s12976-019-0109-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND After proteolysis, the majority of released amino acids from dietary protein are transported to the liver for gluconeogenesis or to peripheral tissues where they are used for protein synthesis and eventually catabolized, producing ammonia as a byproduct. High ammonia levels in the brain are a major contributor to the decreased neural function that occurs in several pathological conditions such as hepatic encephalopathy when liver urea cycle function is compromised. Therefore, it is important to gain a deeper understanding of human ammonia metabolism. The objective of this study was to predict changes in blood ammonia levels resulting from alterations in dietary protein intake, from liver disease, or from partial loss of urea cycle function. METHODS A simple mathematical model was created using MATLAB SimBiology and data from published studies. Simulations were performed and results analyzed to determine steady state changes in ammonia levels resulting from varying dietary protein intake and varying liver enzyme activity levels to simulate liver disease. As a toxicity reference, viability was measured in SH-SY5Y neuroblastoma cells following differentiation and ammonium chloride treatment. RESULTS Results from control simulations yielded steady state blood ammonia levels within normal physiological limits. Increasing dietary protein intake by 72% resulted in a 59% increase in blood ammonia levels. Simulations of liver cirrhosis increased blood ammonia levels by 41 to 130% depending upon the level of dietary protein intake. Simulations of heterozygous individuals carrying a loss of function allele of the urea cycle carbamoyl phosphate synthetase I (CPS1) gene resulted in more than a tripling of blood ammonia levels (from roughly 18 to 60 μM depending on dietary protein intake). The viability of differentiated SH-SY5Y cells was decreased by 14% by the addition of a slightly higher amount of ammonium chloride (90 μM). CONCLUSIONS Data from the model suggest decreasing protein consumption may be one simple strategy to decrease blood ammonia levels and minimize the risk of developing hepatic encephalopathy for many liver disease patients. In addition, the model suggests subjects who are known carriers of disease-causing CPS1 alleles may benefit from monitoring blood ammonia levels and limiting the level of protein intake if ammonia levels are high.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN USA
| |
Collapse
|
23
|
Vemula P, Jing Y, Zhang H, Hunt JB, Sandusky‑Beltran LA, Lee DC, Liu P. Altered brain arginine metabolism in a mouse model of tauopathy. Amino Acids 2019; 51:513-528. [PMID: 30604097 PMCID: PMC6431576 DOI: 10.1007/s00726-018-02687-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/12/2018] [Indexed: 12/26/2022]
Abstract
Tauopathies consist of intracellular accumulation of hyperphosphorylated and aggregated microtubule protein tau, which remains a histopathological feature of Alzheimer's disease (AD) and frontotemporal dementia. L-Arginine is a semi-essential amino acid with a number of bioactive molecules. Its downstream metabolites putrescine, spermidine, and spermine (polyamines) are critically involved in microtubule assembly and stabilization. Recent evidence implicates altered arginine metabolism in the pathogenesis of AD. Using high-performance liquid chromatographic and mass spectrometric assays, the present study systematically determined the tissue concentrations of L-arginine and its nine downstream metabolites in the frontal cortex, hippocampus, parahippocampal region, striatum, thalamus, and cerebellum in male PS19 mice-bearing human tau P301S mutation at 4, 8, and 12-14 months of age. As compared to their wild-type littermates, PS19 mice displayed early and/or prolonged increases in L-ornithine and altered polyamine levels with age. There were also genotype- and age-related changes in L-arginine, L-citrulline, glutamine, glutamate, and γ-aminobutyric acid in a region- and/or chemical-specific manner. The results demonstrate altered brain arginine metabolism in PS19 mice with the most striking changes in L-ornithine, polyamines, and glutamate, indicating a shift of L-arginine metabolism to favor the arginase-polyamine pathway. Given the role of polyamines in maintaining microtubule stability, the functional significance of these changes remains to be explored in future research.
Collapse
Affiliation(s)
- Pranav Vemula
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Dunedin, New Zealand
| | - Yu Jing
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Dunedin, New Zealand
| | - Hu Zhang
- School of Pharmacy, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Dunedin, New Zealand
| | - Jerry B. Hunt
- Byrd Alzheimer’s Institute, College of Pharmacy and Pharmaceutical Sciences, University of South Florida, Florida, USA
| | - Leslie A. Sandusky‑Beltran
- Byrd Alzheimer’s Institute, College of Pharmacy and Pharmaceutical Sciences, University of South Florida, Florida, USA
| | - Daniel C. Lee
- Byrd Alzheimer’s Institute, College of Pharmacy and Pharmaceutical Sciences, University of South Florida, Florida, USA
| | - Ping Liu
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- School of Pharmacy, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Dunedin, New Zealand
| |
Collapse
|
24
|
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
|
25
|
Abstract
Various fungi and bacteria can colonize in the brain and produce physical alterations seen in Alzheimer’s disease (AD). Environmental and genetic factors affect the occurrence of fungal colonization, and how fungi can grow, enter the brain, and interact with the innate immune system. The essence of AD development is the defeat of the innate immune system, whether through vulnerable patient health status or treatment that suppresses inflammation by suppressing the innate immune system. External and mechanical factors that lead to inflammation are a door for pathogenic opportunity. Current research associates the presence of fungi in the etiology of AD and is shown in cerebral tissue at autopsy. From the time of the discovery of AD, much speculation exists for an infective cause. Identifying any AD disease organism is obscured by processes that can take place over years. Amyloid protein deposits are generally considered to be evidence of an intrinsic response to stress or imbalance, but instead amyloid may be evidence of the innate immune response which exists to destroy fungal colonization through structural interference and cytotoxicity. Fungi can remain ensconced for a long time in niches or inside cells, and it is the harboring of fungi that leads to repeated reinfection and slow wider colonization that eventually leads to a grave outcome. Although many fungi and bacteria are associated with AD affected tissues, discussion here focuses on Candida albicans as the archetype of human fungal pathology because of its wide proliferation as a commensal fungus, extensive published research, numerous fungal morphologies, and majority proliferation in AD tissues.
Collapse
Affiliation(s)
- Bodo Parady
- Children's Hospital Oakland Research Institute, Oakland, CA, USA.,Visiting Scholar, University of California, Berkeley, Berkeley CA, USA
| |
Collapse
|
26
|
Abstract
Brain iron is tightly regulated by a multitude of proteins to ensure homeostasis. Iron dyshomeostasis has become a molecular signature associated with aging which is accompanied by progressive decline in cognitive processes. A common theme in neurodegenerative diseases where age is the major risk factor, iron dyshomeostasis coincides with neuroinflammation, abnormal protein aggregation, neurodegeneration, and neurobehavioral deficits. There is a great need to determine the mechanisms governing perturbations in iron metabolism, in particular to distinguish between physiological and pathological aging to generate fruitful therapeutic targets for neurodegenerative diseases. The aim of the present review is to focus on the age-related alterations in brain iron metabolism from a cellular and molecular biology perspective, alongside genetics, and neuroimaging aspects in man and rodent models, with respect to normal aging and neurodegeneration. In particular, the relationship between iron dyshomeostasis and neuroinflammation will be evaluated, as well as the effects of systemic iron overload on the brain. Based on the evidence discussed here, we suggest a synergistic use of iron-chelators and anti-inflammatories as putative anti-brain aging therapies to counteract pathological aging in neurodegenerative diseases.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| | - Maryam Clark
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Po-Wah So
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| |
Collapse
|
27
|
Griffin JWD, Liu Y, Bradshaw PC, Wang K. In Silico Preliminary Association of Ammonia Metabolism Genes GLS, CPS1, and GLUL with Risk of Alzheimer's Disease, Major Depressive Disorder, and Type 2 Diabetes. J Mol Neurosci 2018; 64:385-396. [PMID: 29441491 DOI: 10.1007/s12031-018-1035-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/31/2018] [Indexed: 12/28/2022]
Abstract
Ammonia is a toxic by-product of protein catabolism and is involved in changes in glutamate metabolism. Therefore, ammonia metabolism genes may link a range of diseases involving glutamate signaling such as Alzheimer's disease (AD), major depressive disorder (MDD), and type 2 diabetes (T2D). We analyzed data from a National Institute on Aging study with a family-based design to determine if 45 single nucleotide polymorphisms (SNPs) in glutaminase (GLS), carbamoyl phosphate synthetase 1 (CPS1), or glutamate-ammonia ligase (GLUL) genes were associated with AD, MDD, or T2D using PLINK software. HAPLOVIEW software was used to calculate linkage disequilibrium measures for the SNPs. Next, we analyzed the associated variations for potential effects on transcriptional control sites to identify possible functional effects of the SNPs. Of the SNPs that passed the quality control tests, four SNPs in the GLS gene were significantly associated with AD, two SNPs in the GLS gene were associated with T2D, and one SNP in the GLUL gene and three SNPs in the CPS1 gene were associated with MDD before Bonferroni correction. The in silico bioinformatic analysis suggested probable functional roles for six associated SNPs. Glutamate signaling pathways have been implicated in all these diseases, and other studies have detected similar brain pathologies such as cortical thinning in AD, MDD, and T2D. Taken together, these data potentially link GLS with AD, GLS with T2D, and CPS1 and GLUL with MDD and stimulate the generation of testable hypotheses that may help explain the molecular basis of pathologies shared by these disorders.
Collapse
Affiliation(s)
- Jeddidiah W D Griffin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| | - Ying Liu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Patrick C Bradshaw
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Kesheng Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
28
|
Amino Acid Catabolism in Alzheimer's Disease Brain: Friend or Foe? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5472792. [PMID: 28261376 PMCID: PMC5316456 DOI: 10.1155/2017/5472792] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/04/2016] [Accepted: 01/04/2017] [Indexed: 01/08/2023]
Abstract
There is a dire need to discover new targets for Alzheimer's disease (AD) drug development. Decreased neuronal glucose metabolism that occurs in AD brain could play a central role in disease progression. Little is known about the compensatory neuronal changes that occur to attempt to maintain energy homeostasis. In this review using the PubMed literature database, we summarize evidence that amino acid oxidation can temporarily compensate for the decreased glucose metabolism, but eventually altered amino acid and amino acid catabolite levels likely lead to toxicities contributing to AD progression. Because amino acids are involved in so many cellular metabolic and signaling pathways, the effects of altered amino acid metabolism in AD brain are far-reaching. Possible pathological results from changes in the levels of several important amino acids are discussed. Urea cycle function may be induced in endothelial cells of AD patient brains, possibly to remove excess ammonia produced from increased amino acid catabolism. Studying AD from a metabolic perspective provides new insights into AD pathogenesis and may lead to the discovery of dietary metabolite supplements that can partially compensate for alterations of enzymatic function to delay AD or alleviate some of the suffering caused by the disease.
Collapse
|
29
|
Das S, Pal S, Mitra M. Significance of Exhaled Breath Test in Clinical Diagnosis: A Special Focus on the Detection of Diabetes Mellitus. J Med Biol Eng 2016; 36:605-624. [PMID: 27853412 PMCID: PMC5083779 DOI: 10.1007/s40846-016-0164-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Abstract
Analysis of volatile organic compounds (VOCs) emanating from human exhaled breath can provide deep insight into the status of various biochemical processes in the human body. VOCs can serve as potential biomarkers of physiological and pathophysiological conditions related to several diseases. Breath VOC analysis, a noninvasive and quick biomonitoring approach, also has potential for the early detection and progress monitoring of several diseases. This paper gives an overview of the major VOCs present in human exhaled breath, possible biochemical pathways of breath VOC generation, diagnostic importance of their analysis, and analytical techniques used in the breath test. Breath analysis relating to diabetes mellitus and its characteristic breath biomarkers is focused on. Finally, some challenges and limitations of the breath test are discussed.
Collapse
Affiliation(s)
- Souvik Das
- Department of Biomedical Engineering, JIS College of Engineering, Kalyani, West Bengal 741235 India
| | - Saurabh Pal
- Department of Applied Physics, University of Calcutta, Kolkata, West Bengal 700009 India
| | - Madhuchhanda Mitra
- Department of Applied Physics, University of Calcutta, Kolkata, West Bengal 700009 India
| |
Collapse
|
30
|
Adlimoghaddam A, Sabbir MG, Albensi BC. Ammonia as a Potential Neurotoxic Factor in Alzheimer's Disease. Front Mol Neurosci 2016; 9:57. [PMID: 27551259 PMCID: PMC4976099 DOI: 10.3389/fnmol.2016.00057] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/06/2016] [Indexed: 11/13/2022] Open
Abstract
Ammonia is known to be a potent neurotoxin that causes severe negative effects on the central nervous system. Excessive ammonia levels have been detected in the brain of patients with neurological disorders such as Alzheimer disease (AD). Therefore, ammonia could be a factor contributing to the progression of AD. In this review, we provide an introduction to the toxicity of ammonia and putative ammonia transport proteins. We also hypothesize how ammonia may be linked to AD. Additionally, we discuss the evidence that support the hypothesis that ammonia is a key factor contributing to AD progression. Lastly, we summarize the old and new experimental evidence that focuses on energy metabolism, mitochondrial function, inflammatory responses, excitatory glutamatergic, and GABAergic neurotransmission, and memory in support of our ammonia-related hypotheses of AD.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada
| | - Mohammad G Sabbir
- Division of Neurodegenerative Disorders, St. Boniface Hospital Research Winnipeg, MB, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital ResearchWinnipeg, MB, Canada; Department of Pharmacology & Therapeutics, University of ManitobaWinnipeg, MB, Canada
| |
Collapse
|
31
|
Gonzalez-Riano C, Garcia A, Barbas C. Metabolomics studies in brain tissue: A review. J Pharm Biomed Anal 2016; 130:141-168. [PMID: 27451335 DOI: 10.1016/j.jpba.2016.07.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 12/11/2022]
Abstract
Brain is still an organ with a composition to be discovered but beyond that, mental disorders and especially all diseases that curse with dementia are devastating for the patient, the family and the society. Metabolomics can offer an alternative tool for unveiling new insights in the discovery of new treatments and biomarkers of mental disorders. Until now, most of metabolomic studies have been based on biofluids: serum/plasma or urine, because brain tissue accessibility is limited to animal models or post mortem studies, but even so it is crucial for understanding the pathological processes. Metabolomics studies of brain tissue imply several challenges due to sample extraction, along with brain heterogeneity, sample storage, and sample treatment for a wide coverage of metabolites with a wide range of concentrations of many lipophilic and some polar compounds. In this review, the current analytical practices for target and non-targeted metabolomics are described and discussed with emphasis on critical aspects: sample treatment (quenching, homogenization, filtration, centrifugation and extraction), analytical methods, as well as findings considering the used strategies. Besides that, the altered analytes in the different brain regions have been associated with their corresponding pathways to obtain a global overview of their dysregulation, trying to establish the link between altered biological pathways and pathophysiological conditions.
Collapse
Affiliation(s)
- Carolina Gonzalez-Riano
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte 28668, Madrid, Spain
| | - Antonia Garcia
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte 28668, Madrid, Spain.
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte 28668, Madrid, Spain
| |
Collapse
|
32
|
Carnitine and/or Acetylcarnitine Deficiency as a Cause of Higher Levels of Ammonia. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2920108. [PMID: 26998483 PMCID: PMC4779505 DOI: 10.1155/2016/2920108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/27/2016] [Indexed: 12/24/2022]
Abstract
Blood carnitine and/or acetylcarnitine deficiencies are postulated in the literature as possible causes of higher ammonia levels. The aim of this study was to investigate if the use of valproic acid, the age of the patients, or certain central nervous system pathologies can cause carnitine and/or acetylcarnitine deficiency leading to increased ammonia levels. Three groups of patients were studied: (A) epileptic under phenytoin monotherapy (n = 31); (B) with bipolar disorder under valproic acid treatment (n = 28); (C) elderly (n = 41). Plasma valproic acid and blood carnitine and acyl carnitine profiles were determined using a validated HPLC and LC-MS/MS method, respectively. Blood ammonia concentration was determined using an enzymatic automated assay. Higher ammonia levels were encountered in patients under valproic acid treatment and in the elderly. This may be due to the lower carnitine and/or acetylcarnitine found in these patients. Patients with controlled seizures had normal carnitine and acetylcarnitine levels. Further studies are necessary in order to conclude if the uncontrolled bipolar disorder could be the cause of higher carnitine and/or acetylcarnitine levels.
Collapse
|
33
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a dreadful, devastating and incurable motor neuron disease. Aetiologically, it is a multigenic, multifactorial and multiorgan disease. Despite intense research, ALS pathology remains unexplained. Following extensive literature review, this paper posits a new integrative explanation. This framework proposes that ammonia neurotoxicity is a main player in ALS pathogenesis. According to this explanation, a combination of impaired ammonia removal- mainly because of impaired hepatic urea cycle dysfunction-and increased ammoniagenesis- mainly because of impaired glycolytic metabolism in fast twitch skeletal muscle-causes chronic hyperammonia in ALS. In the absence of neuroprotective calcium binding proteins (calbindin, calreticulin and parvalbumin), elevated ammonia-a neurotoxin-damages motor neurons. Ammonia-induced motor neuron damage occurs through multiple mechanisms such as macroautophagy-endolysosomal impairment, endoplasmic reticulum (ER) stress, CDK5 activation, oxidative/nitrosative stress, neuronal hyperexcitability and neuroinflammation. Furthermore, the regional pattern of calcium binding proteins' loss, owing to either ER stress and/or impaired oxidative metabolism, determines clinical variability of ALS. Most importantly, this new framework can be generalised to explain other neurodegenerative disorders such as Huntington's disease and Parkinsonism.
Collapse
Affiliation(s)
- Bhavin Parekh
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| |
Collapse
|
34
|
Evans AR, Gu L, Guerrero R, Robinson RAS. Global cPILOT analysis of the APP/PS-1 mouse liver proteome. Proteomics Clin Appl 2015; 9:872-84. [PMID: 25620666 DOI: 10.1002/prca.201400149] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/20/2014] [Accepted: 01/21/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE A quantitative proteomics strategy called combined precursor isotopic labeling and isobaric tagging (cPILOT) was designed to discover alterations in the amyloid precursor protein/presenilin-1 (APP/PS-1) mouse liver proteome. The multiplexing strategy allows simultaneous quantitation of 12 samples in a single experiment. EXPERIMENTAL DESIGN For cPILOT samples, six APP/PS-1 and six heterozygous mouse livers were modified using precursor dimethylation (pH 2.5) followed by isobaric tagging (pH 8.0). Samples were pooled, fractioned with strong cation exchange, and analyzed using RPLC-MS(3) for protein identification and relative quantitation. In order to increase proteome coverage, a two-tiered data collection strategy was employed. Six duplex precursor dimethylation experiments were also performed to verify cPILOT protein quantitation. RESULTS The combination of cPILOT with precursor dimethylation data resulted in 2437 total liver proteins identified and 77 differentially expressed proteins in APP/PS-1 liver. Differentially expressed proteins are involved in metabolic processes such as B-oxidation, pyruvate metabolism, and glucose regulation. CONCLUSIONS AND CLINICAL RELEVANCE cPILOT expands protein quantitation using isobaric tags and can be applied to any clinical laboratory interested in enhanced multiplexing strategies. Differentially expressed proteins in APP/PS-1 mouse liver suggest the potential use of ketone bodies to alleviate metabolic dysregulation in Alzheimer's disease brain. Our work also suggests alterations in the alanine cycle potentially leading to hyperammonia production, may contribute to Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Adam R Evans
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Liqing Gu
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rodolfo Guerrero
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Renã A S Robinson
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
35
|
González-Domínguez R, García-Barrera T, Vitorica J, Gómez-Ariza JL. High throughput multiorgan metabolomics in the APP/PS1 mouse model of Alzheimer's disease. Electrophoresis 2015; 36:2237-2249. [PMID: 25641566 DOI: 10.1002/elps.201400544] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 12/29/2022]
Abstract
Metabolomics has demonstrated a great potential for the study of pathological mechanisms occurring in brain from Alzheimer's disease patients and transgenic models. However, its application to peripheral samples is not so common, although it can provide interesting information about systemic abnormalities underlying to disease. This work represents the first metabolomic investigation of multiple peripheral organs (liver, kidney, spleen, and thymus) from the APP/PS1 mice by using a high-throughput approach based on direct infusion MS. Our findings demonstrated that these organs suffer significant metabolic impairments related to energy metabolism (e.g. glycolysis, Krebs cycle, β-oxidation), lipid homeostasis (e.g. cellular membrane breakdown and fatty acid metabolism), degradation of nucleotides, oxidative stress, hyperammonemia, and metabolism of amino acids. It is noteworthy that many of these alterations have been previously described in brain, confirming the systemic character of this neurodegenerative disorder and the utility of peripheral samples to understand its pathogenesis.
Collapse
Affiliation(s)
- Raúl González-Domínguez
- Department of Chemistry and CC.MM, Faculty of Experimental Sciences, University of Huelva, Campus de El Carmen, Huelva, Spain.,Campus of Excellence International ceiA3, University of Huelva, Huelva, Spain.,Research Center of Health and Environment (CYSMA), Campus de El Carmen, University of Huelva, Huelva, Spain
| | - Tamara García-Barrera
- Department of Chemistry and CC.MM, Faculty of Experimental Sciences, University of Huelva, Campus de El Carmen, Huelva, Spain.,Campus of Excellence International ceiA3, University of Huelva, Huelva, Spain.,Research Center of Health and Environment (CYSMA), Campus de El Carmen, University of Huelva, Huelva, Spain
| | - Javier Vitorica
- Department Bioquímica, Bromatologia, Toxicología y Medicina Legal, Faculty of Pharmacy, University of Seville, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Seville, Spain.,Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío, CSIC, University of Seville, Seville, Spain
| | - José Luis Gómez-Ariza
- Department of Chemistry and CC.MM, Faculty of Experimental Sciences, University of Huelva, Campus de El Carmen, Huelva, Spain.,Campus of Excellence International ceiA3, University of Huelva, Huelva, Spain.,Research Center of Health and Environment (CYSMA), Campus de El Carmen, University of Huelva, Huelva, Spain
| |
Collapse
|
36
|
Samsel A, Seneff S. Glyphosate, pathways to modern diseases III: Manganese, neurological diseases, and associated pathologies. Surg Neurol Int 2015; 6:45. [PMID: 25883837 PMCID: PMC4392553 DOI: 10.4103/2152-7806.153876] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/21/2015] [Indexed: 12/20/2022] Open
Abstract
Manganese (Mn) is an often overlooked but important nutrient, required in small amounts for multiple essential functions in the body. A recent study on cows fed genetically modified Roundup(®)-Ready feed revealed a severe depletion of serum Mn. Glyphosate, the active ingredient in Roundup(®), has also been shown to severely deplete Mn levels in plants. Here, we investigate the impact of Mn on physiology, and its association with gut dysbiosis as well as neuropathologies such as autism, Alzheimer's disease (AD), depression, anxiety syndrome, Parkinson's disease (PD), and prion diseases. Glutamate overexpression in the brain in association with autism, AD, and other neurological diseases can be explained by Mn deficiency. Mn superoxide dismutase protects mitochondria from oxidative damage, and mitochondrial dysfunction is a key feature of autism and Alzheimer's. Chondroitin sulfate synthesis depends on Mn, and its deficiency leads to osteoporosis and osteomalacia. Lactobacillus, depleted in autism, depend critically on Mn for antioxidant protection. Lactobacillus probiotics can treat anxiety, which is a comorbidity of autism and chronic fatigue syndrome. Reduced gut Lactobacillus leads to overgrowth of the pathogen, Salmonella, which is resistant to glyphosate toxicity, and Mn plays a role here as well. Sperm motility depends on Mn, and this may partially explain increased rates of infertility and birth defects. We further reason that, under conditions of adequate Mn in the diet, glyphosate, through its disruption of bile acid homeostasis, ironically promotes toxic accumulation of Mn in the brainstem, leading to conditions such as PD and prion diseases.
Collapse
Affiliation(s)
- Anthony Samsel
- Research Scientist and Consultant, Deerfield, NH 03037, USA
| | - Stephanie Seneff
- Spoken Language Systems Group, Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge MA 02139, USA
| |
Collapse
|
37
|
Solé M, Miñano-Molina AJ, Unzeta M. A cross-talk between Aβ and endothelial SSAO/VAP-1 accelerates vascular damage and Aβ aggregation related to CAA-AD. Neurobiol Aging 2015; 36:762-75. [DOI: 10.1016/j.neurobiolaging.2014.09.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 09/18/2014] [Accepted: 09/29/2014] [Indexed: 02/07/2023]
|
38
|
Liu P, Fleete MS, Jing Y, Collie ND, Curtis MA, Waldvogel HJ, Faull RLM, Abraham WC, Zhang H. Altered arginine metabolism in Alzheimer's disease brains. Neurobiol Aging 2014; 35:1992-2003. [PMID: 24746363 DOI: 10.1016/j.neurobiolaging.2014.03.013] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 02/27/2014] [Accepted: 03/14/2014] [Indexed: 12/21/2022]
Abstract
L-arginine is a semi-essential amino acid with a number of bioactive metabolites. Accumulating evidence suggests the implication of altered arginine metabolism in the pathogenesis of Alzheimer's disease (AD). The present study systematically compared the metabolic profile of L-arginine in the superior frontal gyrus, hippocampus, and cerebellum from AD (mean age 80 years) and normal (mean age 80 or 60 years) cases. The activity and protein expression of nitric oxide synthase and arginase were altered with AD and age in a region-specific manner. There were also AD- and age-related changes in the tissue concentrations of L-arginine and its downstream metabolites (L-citrulline, L-ornithine, agmatine, putrescine, spermidine, spermine, glutamate, γ-aminobutyric acid, and glutamine) in a metabolite- or region-specific manner. These findings demonstrate that arginine metabolism is dramatically altered in diverse regions of AD brains, thus meriting further investigation to understand its role in the pathogenesis and/or progression of the disease.
Collapse
Affiliation(s)
- Ping Liu
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
| | - Michael S Fleete
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Yu Jing
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Nicola D Collie
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research and Department of Anatomy with Radiology, University of Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research and Department of Anatomy with Radiology, University of Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy with Radiology, University of Auckland, New Zealand
| | - Wickliffe C Abraham
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand; Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Hu Zhang
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand; School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
39
|
Salkovic-Petrisic M, Osmanovic-Barilar J, Knezovic A, Hoyer S, Mosetter K, Reutter W. Long-term oral galactose treatment prevents cognitive deficits in male Wistar rats treated intracerebroventricularly with streptozotocin. Neuropharmacology 2013; 77:68-80. [PMID: 24055495 DOI: 10.1016/j.neuropharm.2013.09.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 08/31/2013] [Accepted: 09/05/2013] [Indexed: 01/02/2023]
Abstract
Basic and clinical research has demonstrated that dementia of sporadic Alzheimer's disease (sAD) type is associated with dysfunction of the insulin-receptor (IR) system followed by decreased glucose transport via glucose transporter GLUT4 and decreased glucose metabolism in brain cells. An alternative source of energy is d-galactose (the C-4-epimer of d-glucose) which is transported into the brain by insulin-independent GLUT3 transporter where it might be metabolized to glucose via the Leloir pathway. Exclusively parenteral daily injections of galactose induce memory deterioration in rodents and are used to generate animal aging model, but the effects of oral galactose treatment on cognitive functions have never been tested. We have investigated the effects of continuous daily oral galactose (200 mg/kg/day) treatment on cognitive deficits in streptozotocin-induced (STZ-icv) rat model of sAD, tested by Morris Water Maze and Passive Avoidance test, respectively. One month of oral galactose treatment initiated immediately after the STZ-icv administration, successfully prevented development of the STZ-icv-induced cognitive deficits. Beneficial effect of oral galactose was independent of the rat age and of the galactose dose ranging from 100 to 300 mg/kg/day. Additionally, oral galactose administration led to the appearance of galactose in the blood. The increase of galactose concentration in the cerebrospinal fluid was several times lower after oral than after parenteral administration of the same galactose dose. Oral galactose exposure might have beneficial effects on learning and memory ability and could be worth investigating for improvement of cognitive deficits associated with glucose hypometabolism in AD.
Collapse
Affiliation(s)
- Melita Salkovic-Petrisic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia.
| | - Jelena Osmanovic-Barilar
- Department of Pharmacology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Siegfried Hoyer
- Department of Pathology, University Clinic, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Kurt Mosetter
- Center for Interdisciplinary Therapies, Obere Laube 44, D-78462 Konstanz, Germany
| | - Werner Reutter
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité, D-14195 Berlin-Dahlem, Germany
| |
Collapse
|
40
|
Vazquez A. Metabolic states following accumulation of intracellular aggregates: implications for neurodegenerative diseases. PLoS One 2013; 8:e63822. [PMID: 23667676 PMCID: PMC3646825 DOI: 10.1371/journal.pone.0063822] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 04/09/2013] [Indexed: 11/18/2022] Open
Abstract
The formation of intracellular aggregates is a common etiology of several neurodegenerative diseases. Mitochondrial defects and oxidative stress has been pointed as the major mechanistic links between the accumulation of intracellular aggregates and cell death. In this work we propose a “metabolic cell death by overcrowding” as an alternative hypothesis. Using a model of neuron metabolism, we predict that as the concentration of protein aggregates increases the neurons transit through three different metabolic phases. The first phase (0–6 mM) corresponds with the normal neuron state, where the neuronal activity is sustained by the oxidative phosphorylation of lactate. The second phase (6–8.6 mM) is characterized by a mixed utilization of lactate and glucose as energy substrates and a switch from ammonia uptake to ammonia release by neurons. In the third phase (8.6–9.3 mM) neurons are predicted to support their energy demands from glycolysis and an alternative pathway for energy generation, involving reactions from serine synthesis, one carbon metabolism and the glycine cleavage system. The model also predicts a decrease in the maximum neuronal capacity for energy generation with increasing the concentration of protein aggregates. Ultimately this maximum capacity becomes zero when the protein aggregates reach a concentration of about 9.3 mM, predicting the cessation of neuronal activity.
Collapse
Affiliation(s)
- Alexei Vazquez
- Department of Radiation Oncology and Center for Systems Biology, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, New Jersey, United States of America.
| |
Collapse
|
41
|
Glyphosate’s Suppression of Cytochrome P450 Enzymes and Amino Acid Biosynthesis by the Gut Microbiome: Pathways to Modern Diseases. ENTROPY 2013. [DOI: 10.3390/e15041416] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Felipo V, Urios A, Montesinos E, Molina I, Garcia-Torres ML, Civera M, Olmo JAD, Ortega J, Martinez-Valls J, Serra MA, Cassinello N, Wassel A, Jordá E, Montoliu C. Contribution of hyperammonemia and inflammatory factors to cognitive impairment in minimal hepatic encephalopathy. Metab Brain Dis 2012; 27:51-8. [PMID: 22072427 DOI: 10.1007/s11011-011-9269-3] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 10/25/2011] [Indexed: 12/15/2022]
Abstract
To assess the contribution of hyperammonemia and inflammation to induction of mild cognitive impairment (or MHE). We analyzed the presence of mild cognitive impairment (CI) by using the PHES battery of psychometric tests and measured the levels of ammonia and of the inflammatory cytokines IL-6 and IL-18 in blood of patients with different types of liver or dermatological diseases resulting in different grades of hyperammonemia and/or inflammation. The study included patients with 1) liver cirrhosis, showing hyperammonemia and inflammation; 2) non-alcoholic fatty liver disease (NAFLD) showing inflammation but not hyperammonemia; 3) non-alcoholic steatohepatitis (NASH) showing inflammation and very mild hyperammonemia; 4) psoriasis, showing inflammation but not hyperammonemia; 5) keloids, showing both inflammation and hyperammonemia and 6) controls without inflammation or hyperammonemia. The data reported show that in patients with liver diseases, cognitive impairment may appear before progression to cirrhosis if hyperammonemia and inflammation are high enough. Five out of 11 patients with NASH, without liver cirrhosis, showed cognitive impairment associated with hyperammonemia and inflammation. Patients with keloids showed cognitive impairment associated with hyperammonemia and inflammation, in the absence of liver disease. Hyperammonemia or inflammation alone did not induce CI but the combination of certain levels of hyperammonemia and inflammation is enough to induce CI, even without liver disease.
Collapse
Affiliation(s)
- Vicente Felipo
- Laboratory of Neurobiology. Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhang Q, Meng Z. The inotropic effects of ammonia on isolated perfused rat hearts and the mechanisms involved. ACTA ACUST UNITED AC 2012; 214:4048-54. [PMID: 22071197 DOI: 10.1242/jeb.055947] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ammonia (NH(3)) is a common exogenous gas in the atmosphere, as well as an endogenous chemical produced by amino acid catabolism and other pathways in vivo. Physiological and pathophysiological roles of NH(3) in the nervous system have been studied. Recently, endogenous NH(3) has been suggested to be a gas transmitter. However, so far the role of NH(3) in cardiovascular functions has not been reported. The present study was designed to investigate the inotropic effects of NH(3) on isolated perfused rat hearts and the possible mechanisms involved in these effects. The results showed that NH(3) had a positive inotropic effect in a concentration-dependent manner and produced a higher positive effect than NaOH and NH(4)Cl. At low concentrations, the effect of NH(3) on cardiac function was caused by NH(3) molecules; at high concentrations, the effect of NH(3) on hearts may be partly correlated with a change of pH value, but was mainly caused by NH(3) molecules. The mechanisms involved in the NH(3)-induced positive inotropic effect may be related to the ATP-sensitive K(+) (K(ATP)) channel and the nitric oxide (NO)-cyclic GMP (cGMP) signaling pathway. In addition, at a concentration of 1.5 mmol l(-1), NH(3) significantly increased the activity of creatine kinase (CK) and lactate dehydrogenase (LDH) in the coronary perfusate and decreased the activity of Na(+),K(+)-ATPase and Ca(2+),Mg(2+)-ATPase in the hearts. These results indicate that NH(3) at physiological or low concentrations may play a modulatory role in heart function, but at high concentrations had a damaging effect on isolated rat hearts.
Collapse
Affiliation(s)
- Quanxi Zhang
- Institute of Environmental Medicine and Toxicology, Research Center of Environmental Science and Engineering, Shanxi University, Taiyuan 030006, China
| | | |
Collapse
|
44
|
Contribution of hyperammonemia and inflammatory factors to cognitive impairment in minimal hepatic encephalopathy. Metab Brain Dis 2011. [PMID: 22072427 DOI: 10.1007/s11011-011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To assess the contribution of hyperammonemia and inflammation to induction of mild cognitive impairment (or MHE). We analyzed the presence of mild cognitive impairment (CI) by using the PHES battery of psychometric tests and measured the levels of ammonia and of the inflammatory cytokines IL-6 and IL-18 in blood of patients with different types of liver or dermatological diseases resulting in different grades of hyperammonemia and/or inflammation. The study included patients with 1) liver cirrhosis, showing hyperammonemia and inflammation; 2) non-alcoholic fatty liver disease (NAFLD) showing inflammation but not hyperammonemia; 3) non-alcoholic steatohepatitis (NASH) showing inflammation and very mild hyperammonemia; 4) psoriasis, showing inflammation but not hyperammonemia; 5) keloids, showing both inflammation and hyperammonemia and 6) controls without inflammation or hyperammonemia. The data reported show that in patients with liver diseases, cognitive impairment may appear before progression to cirrhosis if hyperammonemia and inflammation are high enough. Five out of 11 patients with NASH, without liver cirrhosis, showed cognitive impairment associated with hyperammonemia and inflammation. Patients with keloids showed cognitive impairment associated with hyperammonemia and inflammation, in the absence of liver disease. Hyperammonemia or inflammation alone did not induce CI but the combination of certain levels of hyperammonemia and inflammation is enough to induce CI, even without liver disease.
Collapse
|
45
|
Strömberg N, Hakonen A. Plasmophore sensitized imaging of ammonia release from biological tissues using optodes. Anal Chim Acta 2011; 704:139-45. [PMID: 21907031 DOI: 10.1016/j.aca.2011.08.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 08/02/2011] [Accepted: 08/08/2011] [Indexed: 11/26/2022]
Abstract
A plasmophore sensitized optode was developed for imaging ammonia (NH(3)) concentrations in muscle tissues. The developed ammonia sensor and an equivalent non plasmophore version of the sensor were tested side by side to compare their limit of detection, dynamic range, reversibility and overall imaging quality. Bio-degradation patterns of ammonia release from lean porcine skeletal muscle were studied over a period of 11 days. We demonstrate that ammonia concentrations ranging from 10nM can be quantified reversibly with an optical resolution of 127 μm in a sample area of 25 mm × 35 mm. The plasmophore ammonia optode showed improved reversibility, less false pixels and a 2 nM ammonia detection limit compared to 200 nM for the non-plasmophore sensor. Main principles of the sensing mechanism include ammonia transfer over a gas permeable film, ammonia protonation, nonactin facilitated merocyanine-ammonium coextraction and plasmophore enhancement. The vast signal improvement is suggested to rely on solvatochroism, nanoparticle scattering and plasmonic interactions that are utilized constructively in a fluorescence ratio. In addition to fundamental medicinal and biological research applications in tissue physiology, reversible ammonia quantification will be possible for a majority of demanding imaging and non imaging applications such as monitoring of low ammonia background concentrations in air and non-invasive medicinal diagnosis through medical breath or saliva analysis. The nanoparticle doped sensor constitutes a highly competitive technique for ammonia sensing in complex matrixes and the general sensing scheme offers new possibilities for the development of artificial optical noses and tongues.
Collapse
|
46
|
Weston CJ, Adams DH. Hepatic consequences of vascular adhesion protein-1 expression. J Neural Transm (Vienna) 2011; 118:1055-64. [PMID: 21512782 DOI: 10.1007/s00702-011-0647-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 04/10/2011] [Indexed: 01/09/2023]
|
47
|
Hansmannel F, Sillaire A, Kamboh MI, Lendon C, Pasquier F, Hannequin D, Laumet G, Mounier A, Ayral AM, DeKosky ST, Hauw JJ, Berr C, Mann D, Amouyel P, Campion D, Lambert JC. Is the urea cycle involved in Alzheimer's disease? J Alzheimers Dis 2011; 21:1013-21. [PMID: 20693631 DOI: 10.3233/jad-2010-100630] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Since previous observations indicated that the urea cycle may have a role in the Alzheimer's disease (AD) process, we set out to quantify the expression of each gene involved in the urea cycle in control and AD brains and establish whether these genes could be genetic determinants of AD. We first confirmed that all the urea cycle enzyme genes are expressed in the AD brain. The expression of arginase 2 was greater in the AD brain than in the control brain. The presence of the rare arginase 2 allele rs742869 was associated with an increase in the risk of AD in men and with an earlier age-at-onset for both genders. None of the other genes in the pathway appeared to be differentially expressed in the AD brain or act as genetic determinants of the disease.
Collapse
|
48
|
Reactive astrocytes give neurons less support: implications for Alzheimer's disease. Neurobiol Aging 2010; 33:423.e1-13. [PMID: 21051108 DOI: 10.1016/j.neurobiolaging.2010.09.018] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 09/20/2010] [Accepted: 09/20/2010] [Indexed: 11/20/2022]
Abstract
Astrocytes become activated in Alzheimer's disease (AD), contributing to and reinforcing an inflammatory cascade. It is proposed that by transforming from a basal to a reactive state, astrocytes neglect their neurosupportive functions, thus rendering neurons vulnerable to excitotoxicity and oxidative stress. This review considers 3 important astrocytic functions, that when disrupted, can affect neuronal metabolism. These are the uptake of glucose and release of lactate; the uptake of glutamate and release of glutamine; and the uptake of glutathione precursors and release of glutathione. Conditions under which these functions can be manipulated in vitro, as well as examples of possible loss of astrocytic function in AD, are discussed. It is proposed that the targeting of astrocytes with pharmacological agents that are specifically designed to return astrocytes to a quiescent phenotype could represent a fruitful new angle for the therapeutic treatment of AD and other neurodegenerative disorders.
Collapse
|
49
|
Kaminsky Y, Kosenko E. AMP deaminase and adenosine deaminase activities in liver and brain regions in acute ammonia intoxication and subacute toxic hepatitis. Brain Res 2009; 1311:175-81. [PMID: 19900420 DOI: 10.1016/j.brainres.2009.10.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 10/29/2009] [Accepted: 10/30/2009] [Indexed: 10/20/2022]
Abstract
Cytosolic enzymes AMP deaminase and adenosine deaminase (ADA) catalyze AMP and adenosine deamination, constitute rate-limiting steps of adenine nucleotide catabolism and play important roles in cellular energy metabolism. In this study, AMP deaminase and ADA activities of rat liver, neocortex, cerebellum, striatum and hippocampus were investigated in acute ammonia intoxication and subacute CCl(4)-induced hepatitis. Activities of both AMP deaminase and ADA in the liver were elevated by 2.4-4.2-fold (p<0.0001) in both models of hepatotoxic injury as compared with controls. In acute hyperammonemia activities of AMP, deaminase and ADA increased by 46-59% (p<0.02) in the neocortex and did not change in the striatum. In the hippocampus of hyperammonemic rats, only AMP deaminase activity was increased by 48% (p=0.0004), and in the cerebellum only ADA activity was increased significantly (by 26%, p<0.05). The adenylate pool size and energy charge were greatly reduced in the neocortex of hyperammonemic rats. Results suggested that two parallel pathways of AMP breakdown, including AMP deaminase and ADA, respectively, are up-regulated under pathological conditions, probably in order to overcome compensatory synthesis of adenylates, to ensure prompt adenylate pool depletion and reduce the adenylate energy charge in liver and selected brain regions.
Collapse
Affiliation(s)
- Yury Kaminsky
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia.
| | | |
Collapse
|
50
|
Kaminsky YG, Marlatt MW, Smith MA, Kosenko EA. Subcellular and metabolic examination of amyloid-beta peptides in Alzheimer disease pathogenesis: evidence for Abeta(25-35). Exp Neurol 2009; 221:26-37. [PMID: 19751725 DOI: 10.1016/j.expneurol.2009.09.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 09/03/2009] [Accepted: 09/04/2009] [Indexed: 11/28/2022]
Abstract
Amyloid-beta peptide (Abeta) is a central player in the pathogenesis and diagnosis of Alzheimer disease. It aggregates to form the core of Alzheimer disease-associated plaques found in coordination with tau deposits in diseased individuals. Despite this clinical relevance, no single hypothesis satisfies and explicates the role of Abeta in toxicity and progression of the disease. To explore this area, investigators have focused on mechanisms of cellular dysfunction, aggregation, and maladaptive responses. Extensive research has been conducted using various methodologies to investigate Abeta peptides and oligomers, and these multiple facets have provided a wealth of data from specific models. Notably, the utility of each experiment must be considered in regards to the brain environment. The use of Abeta(25-35) in studies of cellular dysfunction has provided data indicating that the peptide is indeed responsible for multiple disturbances to cellular integrity. We will review how Abeta peptide induces oxidative stress and calcium homeostasis, and how multiple enzymes are deleteriously impacted by Abeta(25-35). Understanding and discussing the origin and properties of Abeta peptides is essential to evaluating their effects on various intracellular metabolic processes. Attention will also be specifically directed to metabolic compartmentation in affected brain cells, including mitochondrial, cytosolic, nuclear, and lysosomal enzymes.
Collapse
Affiliation(s)
- Yury G Kaminsky
- Institute of Theoretical and Experimental Biophysics, RAS, Pushchino, Russia.
| | | | | | | |
Collapse
|