1
|
Kalecký K, Buitrago L, Alarcon JM, Singh A, Bottiglieri T, Kaddurah-Daouk R, Hernández AI. Rescue of hippocampal synaptic plasticity and memory performance by Fingolimod (FTY720) in APP/PS1 model of Alzheimer's disease is accompanied by correction in metabolism of sphingolipids, polyamines, and phospholipid saturation composition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633452. [PMID: 39868189 PMCID: PMC11761635 DOI: 10.1101/2025.01.17.633452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Previously, our metabolomic, transcriptomic, and genomic studies characterized the ceramide/sphingomyelin pathway as a therapeutic target in Alzheimer's disease, and we demonstrated that FTY720, a sphingosine-1-phospahate receptor modulator approved for treatment of multiple sclerosis, recovers synaptic plasticity and memory in APP/PS1 mice. To further investigate how FTY720 rescues the pathology, we performed metabolomic analysis in brain, plasma, and liver of trained APP/PS1 and wild-type mice. APP/PS1 mice showed area-specific brain disturbances in polyamines, phospholipids, and sphingolipids. Most changes were completely or partially normalized in FTY720-treated subjects, indicating rebalancing the "sphingolipid rheostat", reactivating phosphatidylethanolamine synthesis via mitochondrial phosphatidylserine decarboxylase pathway, and normalizing polyamine levels that support mitochondrial activity. Synaptic plasticity and memory were rescued, with spermidine synthesis in temporal cortex best corresponding to hippocampal CA3-CA1 plasticity normalization. FTY720 effects, also reflected in other pathways, are consistent with promotion of mitochondrial function, synaptic plasticity, and anti-inflammatory environment, while reducing pro-apoptotic and pro-inflammatory signals.
Collapse
Affiliation(s)
- Karel Kalecký
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Luna Buitrago
- Neural and Behavioral Sciences Program, School of Graduate Studies, Department of Neurology/Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Juan Marcos Alarcon
- Neural and Behavioral Sciences Program, School of Graduate Studies, The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Abanish Singh
- Department of Psychiatry and Behavioral Sciences; and Department of Medicine, Duke University School of Medicine, Durham, Durham, NC, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Alejandro Iván Hernández
- Neural and Behavioral Sciences Program, School of Graduate Studies, The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | | |
Collapse
|
2
|
An C, Cai H, Ren Z, Fu X, Quan S, Jia L. Biofluid biomarkers for Alzheimer's disease: past, present, and future. MEDICAL REVIEW (2021) 2024; 4:467-491. [PMID: 39664082 PMCID: PMC11629312 DOI: 10.1515/mr-2023-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 09/04/2024] [Indexed: 12/13/2024]
Abstract
Alzheimer's disease (AD) is a gradually progressive neurodegenerative disease with tremendous social and economic burden. Therefore, early and accurate diagnosis is imperative for effective treatment or prevention of the disease. Cerebrospinal fluid and blood biomarkers emerge as favorable diagnostic tools due to their relative accessibility and potential for widespread clinical use. This review focuses on the AT(N) biomarker system, which includes biomarkers reflecting AD core pathologies, amyloid deposition, and pathological tau, as well as neurodegeneration. Novel biomarkers associated with inflammation/immunity, synaptic dysfunction, vascular pathology, and α-synucleinopathy, which might contribute to either the pathogenesis or the clinical progression of AD, have also been discussed. Other emerging candidates including non-coding RNAs, metabolites, and extracellular vesicle-based markers have also enriched the biofluid biomarker landscape for AD. Moreover, the review discusses the current challenges of biofluid biomarkers in AD diagnosis and offers insights into the prospective future development.
Collapse
Affiliation(s)
- Chengyu An
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shuiyue Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
3
|
Jie J, Gong Y, Hu H, Liu S. The role of cerebrospinal fluid metabolites in mediating the impact of lipids on Late-Onset Alzheimer's Disease: a two-step mendelian randomization analysis. J Transl Med 2024; 22:1077. [PMID: 39609832 PMCID: PMC11603644 DOI: 10.1186/s12967-024-05796-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Although research has indicated correlations between lipids, cerebrospinal fluid (CSF) metabolites, and Late-Onset Alzheimer's Disease (LOAD), the specific causal relationships among these elements, as well as the roles and mechanisms of the cerebrospinal fluid metabolites, remain unclear. METHODS Statistical datasets derived from Genome-Wide Association Studies (GWAS) were utilized to assess the bidirectional causal relationships between lipids and LOAD. Subsequently, genetic variants associated with CSF metabolites and established lipids underwent a two-step Mendelian randomization (MR) analysis to explore potential mediators and analyze mediation effects. Sensitivity analyses were employed to assess the robustness of the detection systems. RESULTS Genetically predicted cholesterol (IVW OR = 0.989; 95% CI 0.982-0.996) was found to reduce the risk of LOAD, whereas Phosphatidylcholine (PC) (18:1_0:0) (IVW OR = 1.015; 95% CI 1.005-1.025) posed a risk factor. The potential mediator, CSF metabolite N-acetylneuraminate (NeuAC), was identified with a mediation proportion of 21.02% (3.25%, 45.50%). No pleiotropy or heterogeneity was detected across MR analyses. CONCLUSIONS The findings underscore the pivotal role of CSF metabolomics in elucidating the lipid-mediated pathogenesis of LOAD, highlighting potential diagnostic and preventative biomarkers.
Collapse
Affiliation(s)
- Jie Jie
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People' s Hospital of Changde City), 818 Renmin Road, Changde City, Hunan Province, 415000, China
| | - Yonglu Gong
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People' s Hospital of Changde City), 818 Renmin Road, Changde City, Hunan Province, 415000, China
| | - Hongbo Hu
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People' s Hospital of Changde City), 818 Renmin Road, Changde City, Hunan Province, 415000, China
| | - Su Liu
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People' s Hospital of Changde City), 818 Renmin Road, Changde City, Hunan Province, 415000, China.
| |
Collapse
|
4
|
Schweickart A, Batra R, Neth BJ, Martino C, Shenhav L, Zhang AR, Shi P, Karu N, Huynh K, Meikle PJ, Schimmel L, Dilmore AH, Blennow K, Zetterberg H, Blach C, Dorrestein PC, Knight R, Craft S, Kaddurah-Daouk R, Krumsiek J. Serum and CSF metabolomics analysis shows Mediterranean Ketogenic Diet mitigates risk factors of Alzheimer's disease. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:15. [PMID: 38962750 PMCID: PMC11216994 DOI: 10.1038/s44324-024-00016-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/16/2024] [Indexed: 07/05/2024]
Abstract
Alzheimer's disease (AD) is influenced by a variety of modifiable risk factors, including a person's dietary habits. While the ketogenic diet (KD) holds promise in reducing metabolic risks and potentially affecting AD progression, only a few studies have explored KD's metabolic impact, especially on blood and cerebrospinal fluid (CSF). Our study involved participants at risk for AD, either cognitively normal or with mild cognitive impairment. The participants consumed both a modified Mediterranean Ketogenic Diet (MMKD) and the American Heart Association diet (AHAD) for 6 weeks each, separated by a 6-week washout period. We employed nuclear magnetic resonance (NMR)-based metabolomics to profile serum and CSF and metagenomics profiling on fecal samples. While the AHAD induced no notable metabolic changes, MMKD led to significant alterations in both serum and CSF. These changes included improved modifiable risk factors, like increased HDL-C and reduced BMI, reversed serum metabolic disturbances linked to AD such as a microbiome-mediated increase in valine levels, and a reduction in systemic inflammation. Additionally, the MMKD was linked to increased amino acid levels in the CSF, a breakdown of branched-chain amino acids (BCAAs), and decreased valine levels. Importantly, we observed a strong correlation between metabolic changes in the CSF and serum, suggesting a systemic regulation of metabolism. Our findings highlight that MMKD can improve AD-related risk factors, reverse some metabolic disturbances associated with AD, and align metabolic changes across the blood-CSF barrier.
Collapse
Affiliation(s)
- Annalise Schweickart
- Tri-Institutional Program in Computational Biology & Medicine, Weill Cornell Medicine, New York, NY USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, New York, NY USA
| | - Richa Batra
- Department of Physiology and Biophysics, Weill Cornell Medicine, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, New York, NY USA
| | - Bryan J. Neth
- Department of Neurology, Mayo Clinic, Rochester, MN USA
| | - Cameron Martino
- Department of Pediatrics, University of California San Diego, La Jolla, CA USA
| | - Liat Shenhav
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY USA
| | - Anru R. Zhang
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC USA
| | - Pixu Shi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC USA
| | - Naama Karu
- Tasmanian Independent Metabolomics and Analytical Chemistry Solutions (TIMACS), Hobart, TAS Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC Australia
| | - Leyla Schimmel
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC USA
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, NC USA
| | - Pieter C. Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA USA
| | - Rob Knight
- Departments of Pediatrics, Computer Science and Engineering, Bioengineering, University of California San Diego, La Jolla, CA USA
| | - Alzheimer’s Gut Microbiome Project Consortium
- Tri-Institutional Program in Computational Biology & Medicine, Weill Cornell Medicine, New York, NY USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, New York, NY USA
- Department of Neurology, Mayo Clinic, Rochester, MN USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA USA
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC USA
- Tasmanian Independent Metabolomics and Analytical Chemistry Solutions (TIMACS), Hobart, TAS Australia
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC Australia
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC USA
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Duke Molecular Physiology Institute, Duke University, Durham, NC USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA USA
- Departments of Pediatrics, Computer Science and Engineering, Bioengineering, University of California San Diego, La Jolla, CA USA
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston Salem, NC USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC USA
- Department of Medicine, Duke University, Durham, NC USA
| | - Suzanne Craft
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston Salem, NC USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC USA
- Department of Medicine, Duke University, Durham, NC USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Weill Cornell Medicine, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, New York, NY USA
| |
Collapse
|
5
|
Xu D, Dai X, Zhang L, Cai Y, Chen K, Wu J, Dong L, Shen L, Yang J, Zhao J, Zhou Y, Mei Z, Wei W, Zhang Z, Xiong N. Mass spectrometry for biomarkers, disease mechanisms, and drug development in cerebrospinal fluid metabolomics. Trends Analyt Chem 2024; 173:117626. [DOI: 10.1016/j.trac.2024.117626] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
|
6
|
Schweickart A, Batra R, Neth BJ, Martino C, Shenhav L, Zhang AR, Shi P, Karu N, Huynh K, Meikle PJ, Schimmel L, Dilmore AH, Blennow K, Zetterberg H, Blach C, Dorrestein PC, Knight R, Craft S, Kaddurah-Daouk R, Krumsiek J. A Modified Mediterranean Ketogenic Diet mitigates modifiable risk factors of Alzheimer's Disease: a serum and CSF-based metabolic analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.27.23298990. [PMID: 38076824 PMCID: PMC10705656 DOI: 10.1101/2023.11.27.23298990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Alzheimer's disease (AD) is influenced by a variety of modifiable risk factors, including a person's dietary habits. While the ketogenic diet (KD) holds promise in reducing metabolic risks and potentially affecting AD progression, only a few studies have explored KD's metabolic impact, especially on blood and cerebrospinal fluid (CSF). Our study involved participants at risk for AD, either cognitively normal or with mild cognitive impairment. The participants consumed both a modified Mediterranean-ketogenic diet (MMKD) and the American Heart Association diet (AHAD) for 6 weeks each, separated by a 6-week washout period. We employed nuclear magnetic resonance (NMR)-based metabolomics to profile serum and CSF and metagenomics profiling on fecal samples. While the AHAD induced no notable metabolic changes, MMKD led to significant alterations in both serum and CSF. These changes included improved modifiable risk factors, like increased HDL-C and reduced BMI, reversed serum metabolic disturbances linked to AD such as a microbiome-mediated increase in valine levels, and a reduction in systemic inflammation. Additionally, the MMKD was linked to increased amino acid levels in the CSF, a breakdown of branched-chain amino acids (BCAAs), and decreased valine levels. Importantly, we observed a strong correlation between metabolic changes in the CSF and serum, suggesting a systemic regulation of metabolism. Our findings highlight that MMKD can improve AD-related risk factors, reverse some metabolic disturbances associated with AD, and align metabolic changes across the blood-CSF barrier.
Collapse
Affiliation(s)
- Annalise Schweickart
- Tri-Institutional Program in Computational Biology & Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, New York, NY 10021, USA
| | - Richa Batra
- Department of Physiology and Biophysics, Weill Cornell Medicine, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, New York, NY 10021, USA
| | | | - Cameron Martino
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Liat Shenhav
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Anru R. Zhang
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Pixu Shi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Naama Karu
- Tasmanian Independent Metabolomics and Analytical Chemistry Solutions (TIMACS), Hobart, 7008 Tasmania, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - Leyla Schimmel
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | | | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA
| | - Rob Knight
- Departments of Pediatrics, Computer Science and Engineering, Bioengineering, University of California San Diego, La Jolla, CA
| | | | - Suzanne Craft
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Weill Cornell Medicine, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, New York, NY 10021, USA
| |
Collapse
|
7
|
Alam S, Afsar SY, Wolter MA, Volk LM, Mitroi DN, Meyer Zu Heringdorf D, van Echten-Deckert G. S1P Lyase Deficiency in the Brain Promotes Astrogliosis and NLRP3 Inflammasome Activation via Purinergic Signaling. Cells 2023; 12:1844. [PMID: 37508508 PMCID: PMC10378183 DOI: 10.3390/cells12141844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/19/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Astrocytes are critical players in brain health and disease. Brain pathologies and lesions are usually accompanied by astroglial alterations known as reactive astrogliosis. Sphingosine 1-phosphate lyase (SGPL1) catalysis, the final step in sphingolipid catabolism, irreversibly cleaves its substrate sphingosine 1-phosphate (S1P). We have shown that neural ablation of SGPL1 causes accumulation of S1P and hence neuronal damage, cognitive deficits, as well as microglial activation. Moreover, the S1P/S1P-receptor signaling axis enhances ATP production in SGPL1-deficient astrocytes. Using immunohistochemical methods as well as RNA Seq and CUT&Tag we show how S1P signaling causes activation of the astrocytic purinoreceptor P2Y1 (P2Y1R). With specific pharmacological agonists and antagonists, we uncover the P2Y1R as the key player in S1P-induced astrogliosis, and DDX3X mediated the activation of the NLRP3 inflammasome, including caspase-1 and henceforward generation of interleukin-1ß (IL-1ß) and of other proinflammatory cytokines. Our results provide a novel route connecting S1P metabolism and signaling with astrogliosis and the activation of the NLRP3 inflammasome, a central player in neuroinflammation, known to be crucial for the pathogenesis of numerous brain illnesses. Thus, our study opens the door for new therapeutic strategies surrounding S1P metabolism and signaling in the brain.
Collapse
Affiliation(s)
- Shah Alam
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Sumaiya Yasmeen Afsar
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Maya Anik Wolter
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Luisa Michelle Volk
- Institute for General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt am Main, Germany
| | - Daniel Nicolae Mitroi
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institute for General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt am Main, Germany
| | - Gerhild van Echten-Deckert
- LIMES Institute for Membrane Biology and Lipid Biochemistry, Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
8
|
Chung HL, Ye Q, Park YJ, Zuo Z, Mok JW, Kanca O, Tattikota SG, Lu S, Perrimon N, Lee HK, Bellen HJ. Very-long-chain fatty acids induce glial-derived sphingosine-1-phosphate synthesis, secretion, and neuroinflammation. Cell Metab 2023; 35:855-874.e5. [PMID: 37084732 PMCID: PMC10160010 DOI: 10.1016/j.cmet.2023.03.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/10/2023] [Accepted: 03/29/2023] [Indexed: 04/23/2023]
Abstract
VLCFAs (very-long-chain fatty acids) are the most abundant fatty acids in myelin. Hence, during demyelination or aging, glia are exposed to higher levels of VLCFA than normal. We report that glia convert these VLCFA into sphingosine-1-phosphate (S1P) via a glial-specific S1P pathway. Excess S1P causes neuroinflammation, NF-κB activation, and macrophage infiltration into the CNS. Suppressing the function of S1P in fly glia or neurons, or administration of Fingolimod, an S1P receptor antagonist, strongly attenuates the phenotypes caused by excess VLCFAs. In contrast, elevating the VLCFA levels in glia and immune cells exacerbates these phenotypes. Elevated VLCFA and S1P are also toxic in vertebrates based on a mouse model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE). Indeed, reducing VLCFA with bezafibrate ameliorates the phenotypes. Moreover, simultaneous use of bezafibrate and fingolimod synergizes to improve EAE, suggesting that lowering VLCFA and S1P is a treatment avenue for MS.
Collapse
Affiliation(s)
- Hyung-Lok Chung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Qi Ye
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ye-Jin Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jung-Wan Mok
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nobert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Hyun Kyoung Lee
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Hanson AJ, Banks WA, Bettcher LF, Pepin R, Raftery D, Navarro SL, Craft S. Cerebrospinal Fluid Metabolomics: Pilot Study of Using Metabolomics to Assess Diet and Metabolic Interventions in Alzheimer's Disease and Mild Cognitive Impairment. Metabolites 2023; 13:569. [PMID: 37110227 PMCID: PMC10145981 DOI: 10.3390/metabo13040569] [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: 01/18/2023] [Revised: 03/17/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Brain glucose hypometabolism is an early sign of Alzheimer's disease (AD), and interventions which offset this deficit, such as ketogenic diets, show promise as AD therapeutics. Conversely, high-fat feeding may exacerbate AD risk. We analyzed the metabolomic profile of cerebrospinal fluid (CSF) in a pilot study of older adults who underwent saline and triglyceride (TG) infusions. Older adults (12 cognitively normal (CN), age 65.3 ± 8.1, and 9 with cognitive impairment (CI), age 70.9 ± 8.6) underwent a 5 h TG or saline infusion on different days using a random crossover design; CSF was collected at the end of infusion. Aqueous metabolites were measured using a targeted mass spectroscopy (MS) platform focusing on 215 metabolites from over 35 different metabolic pathways. Data were analyzed using MetaboAnalyst 4.0 and SAS. Of the 215 targeted metabolites, 99 were detectable in CSF. Only one metabolite significantly differed by treatment: the ketone body 3-hydroxybutyrate (HBA). Post hoc analyses showed that HBA levels were associated with age and markers of metabolic syndrome and demonstrated different correlation patterns for the two treatments. When analyzed by cognitive diagnosis group, TG-induced increases in HBA were over 3 times higher for those with cognitive impairment (change score CN +9.8 uM ± 8.3, CI +32.4 ± 7.4, p = 0.0191). Interestingly, individuals with cognitive impairment had higher HBA levels after TG infusion than those with normal cognition. These results suggest that interventions that increase plasma ketones may lead to higher brain ketones in groups at risk for AD and should be confirmed in larger intervention studies.
Collapse
Affiliation(s)
- Angela J. Hanson
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - William A. Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98102, USA
| | - Lisa F. Bettcher
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98109, USA
| | - Robert Pepin
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98109, USA
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98109, USA
| | - Sandi L. Navarro
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Suzanne Craft
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27109, USA
| |
Collapse
|
10
|
van Echten-Deckert G. The role of sphingosine 1-phosphate metabolism in brain health and disease. Pharmacol Ther 2023; 244:108381. [PMID: 36907249 DOI: 10.1016/j.pharmthera.2023.108381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.
Collapse
|
11
|
Yin C, Harms AC, Hankemeier T, Kindt A, de Lange ECM. Status of Metabolomic Measurement for Insights in Alzheimer's Disease Progression-What Is Missing? Int J Mol Sci 2023; 24:ijms24054960. [PMID: 36902391 PMCID: PMC10003384 DOI: 10.3390/ijms24054960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease, leading to the progressive loss of memory and other cognitive functions. As there is still no cure for AD, the growth in the number of susceptible individuals represents a major emerging threat to public health. Currently, the pathogenesis and etiology of AD remain poorly understood, while no efficient treatments are available to slow down the degenerative effects of AD. Metabolomics allows the study of biochemical alterations in pathological processes which may be involved in AD progression and to discover new therapeutic targets. In this review, we summarized and analyzed the results from studies on metabolomics analysis performed in biological samples of AD subjects and AD animal models. Then this information was analyzed by using MetaboAnalyst to find the disturbed pathways among different sample types in human and animal models at different disease stages. We discuss the underlying biochemical mechanisms involved, and the extent to which they could impact the specific hallmarks of AD. Then we identify gaps and challenges and provide recommendations for future metabolomics approaches to better understand AD pathogenesis.
Collapse
Affiliation(s)
- Chunyuan Yin
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Amy C. Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Alida Kindt
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Elizabeth C. M. de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Correspondence:
| |
Collapse
|
12
|
Alam S, Afsar SY, Van Echten-Deckert G. S1P Released by SGPL1-Deficient Astrocytes Enhances Astrocytic ATP Production via S1PR 2,4, Thus Keeping Autophagy in Check: Potential Consequences for Brain Health. Int J Mol Sci 2023; 24:ijms24054581. [PMID: 36902011 PMCID: PMC10003137 DOI: 10.3390/ijms24054581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Astrocytes are critical players in brain health and disease. Sphingosine-1-phosphate (S1P), a bioactive signaling lipid, is involved in several vital processes, including cellular proliferation, survival, and migration. It was shown to be crucial for brain development. Its absence is embryonically lethal, affecting, inter alia, the anterior neural tube closure. However, an excess of S1P due to mutations in S1P-lyase (SGPL1), the enzyme responsible for its constitutive removal, is also harmful. Of note, the gene SGPL1 maps to a region prone to mutations in several human cancers and also in S1P-lyase insufficiency syndrome (SPLIS) characterized by several symptoms, including peripheral and central neurological defects. Here, we investigated the impact of S1P on astrocytes in a mouse model with the neural-targeted ablation of SGPL1. We found that SGPL1 deficiency, and hence the accumulation of its substrate, S1P, causes the elevated expression of glycolytic enzymes and preferentially directs pyruvate into the tricarboxylic acid (TCA) cycle through its receptors (S1PR2,4). In addition, the activity of TCA regulatory enzymes was increased, and consequently, so was the cellular ATP content. The high energy load activates the mammalian target of rapamycin (mTOR), thus keeping astrocytic autophagy in check. Possible consequences for the viability of neurons are discussed.
Collapse
|
13
|
Zhang L, Wong LR, Wong P, Shen W, Yang S, Huang L, Lim YA, Ho PCL. Chronic treatment with baicalein alleviates behavioural disorders and improves cerebral blood flow via reverting metabolic abnormalities in a J20 transgenic mouse model of Alzheimer's disease. Brain Behav Immun Health 2023; 28:100599. [PMID: 36817510 PMCID: PMC9931920 DOI: 10.1016/j.bbih.2023.100599] [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: 09/26/2022] [Revised: 01/21/2023] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
Baicalein (BE) has both antioxidant and anti-inflammatory effects. It has also been reported able to improve cerebral blood circulation in brain ischemic injury. However, its chronic efficacy and metabolomics in Alzheimer's disease (AD) remain unknown. In this study, BE at 80 mg/kg was administrated through the oral route in J20 AD transgenic mice aged from aged 4 months to aged 10 months. Metabolic- and neurobehavioural phenotyping was done before and after 6 months' treatment to evaluate the drug efficacy and the relevant mechanisms. Meanwhile, molecular docking was used to study the binding affinity of BE and poly (ADP-ribose) polymerase-1 (PARP-1) which is related to neuronal injury. The open field test showed that BE could suppress hyperactivity in J20 mice and increase the frequency of the target quadrant crossing in the Morris Water Maze test. More importantly, BE restored cerebral blood flow back to the normal level after the chronic treatment. A 1H NMR-based metabolomics study showed that BE treatment could restore the tricarboxylic acid cycle in plasma. And such a treatment could suppress oxidative stress, inhibit neuroinflammation, alleviate mitochondrial dysfunction, improve neurotransmission, and restore amino homeostasis via starch and sucrose metabolism and glycolipid metabolism in the cortex and hippocampus, which could affect the behavioural and cerebral blood flow. These findings showed that BE is a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Li Zhang
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 117583, Singapore,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Ling Rong Wong
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Peiyan Wong
- Neuroscience Phenotyping Core, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Wanxiang Shen
- Department of Chemistry, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Shili Yang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yun-An Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Paul Chi-Lui Ho
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 117583, Singapore,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore,Monash University Malaysia, School of Pharmacy, Subang Jaya, 47500, Selangor, Malaysia,Corresponding author. Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
| |
Collapse
|
14
|
Dysregulation of sphingosine-1-phosphate (S1P) and S1P receptor 1 signaling in the 5xFAD mouse model of Alzheimer's disease. Brain Res 2023; 1799:148171. [PMID: 36410428 DOI: 10.1016/j.brainres.2022.148171] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/22/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Sphingolipid-1-phosphate (S1P) signaling through the activation S1P receptors (S1PRs) plays critical roles in cellular events in the brain. Aberrant S1P metabolism has been identified in the brains of Alzheimer's disease (AD) patients. Our recent studies have shown that treatment with fingolimod, an analog of sphingosine, provides neuroprotective effects in five familiar Alzheimer disease (5xFAD) transgenic mice, resulting in the reduction of amyloid-β (Aβ) neurotoxicity, inhibition of activation of microglia and astrocytes, increased hippocampal neurogenesis, and improved learning and memory. However, the pathways by which dysfunctional S1P and S1PR signaling may associate with the development of AD-like pathology remain unknown. In this study, we investigated the alteration of signaling of S1P/S1P receptor 1 (S1PR1), the most abundant S1PR subtype in the brain, in the cortex of 5xFAD transgenic mice at 3, 8, and 14 months of age. Compared to non-transgenic wildtype (WT) littermates, we found significant decreased levels of sphingosine kinases (SphKs), increased S1P lyase (S1PL), and increased S1PR1 in 8- and 14-month-old, but not in 3-month-old 5xFAD mice. Furthermore, we detected increased activation of the S1PR1 downstream pathway of Akt/mTor/Tau signaling in aging 5xFAD mice. Treatment with fingolimod from 1 to 8 months of age reversed the levels of SphKs, S1PL, and furthermore, those of S1PR1 and its downstream pathway of Akt/mTor/Tau signaling. Together the data reveal that dysregulation of S1P and S1PR signaling may associate with the development of AD-like pathology through Akt/mTor/Tau signaling.
Collapse
|
15
|
Aerqin Q, Wang ZT, Wu KM, He XY, Dong Q, Yu JT. Omics-based biomarkers discovery for Alzheimer's disease. Cell Mol Life Sci 2022; 79:585. [PMID: 36348101 PMCID: PMC11803048 DOI: 10.1007/s00018-022-04614-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorders presenting with the pathological hallmarks of amyloid plaques and tau tangles. Over the past few years, great efforts have been made to explore reliable biomarkers of AD. High-throughput omics are a technology driven by multiple levels of unbiased data to detect the complex etiology of AD, and it provides us with new opportunities to better understand the pathophysiology of AD and thereby identify potential biomarkers. Through revealing the interaction networks between different molecular levels, the ultimate goal of multi-omics is to improve the diagnosis and treatment of AD. In this review, based on the current AD pathology and the current status of AD diagnostic biomarkers, we summarize how genomics, transcriptomics, proteomics and metabolomics are all conducing to the discovery of reliable AD biomarkers that could be developed and used in clinical AD management.
Collapse
Affiliation(s)
- Qiaolifan Aerqin
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Xiao-Yu He
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
16
|
Custodia A, Romaus-Sanjurjo D, Aramburu-Núñez M, Álvarez-Rafael D, Vázquez-Vázquez L, Camino-Castiñeiras J, Leira Y, Pías-Peleteiro JM, Aldrey JM, Sobrino T, Ouro A. Ceramide/Sphingosine 1-Phosphate Axis as a Key Target for Diagnosis and Treatment in Alzheimer's Disease and Other Neurodegenerative Diseases. Int J Mol Sci 2022; 23:8082. [PMID: 35897658 PMCID: PMC9331765 DOI: 10.3390/ijms23158082] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Alzheimer's disease (AD) is considered the most prevalent neurodegenerative disease and the leading cause of dementia worldwide. Sphingolipids, such as ceramide or sphingosine 1-phosphate, are bioactive molecules implicated in structural and signaling functions. Metabolic dysfunction in the highly conserved pathways to produce sphingolipids may lead to or be a consequence of an underlying disease. Recent studies on transcriptomics and sphingolipidomics have observed alterations in sphingolipid metabolism of both enzymes and metabolites involved in their synthesis in several neurodegenerative diseases, including AD. In this review, we highlight the most relevant findings related to ceramide and neurodegeneration, with a special focus on AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tomás Sobrino
- Neuro Aging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINCs), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.C.); (D.R.-S.); (M.A.-N.); (D.Á.-R.); (L.V.-V.); (J.C.-C.); (Y.L.); (J.M.P.-P.); (J.M.A.)
| | - Alberto Ouro
- Neuro Aging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINCs), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.C.); (D.R.-S.); (M.A.-N.); (D.Á.-R.); (L.V.-V.); (J.C.-C.); (Y.L.); (J.M.P.-P.); (J.M.A.)
| |
Collapse
|
17
|
Metabolomics-Based Pharmacodynamic Analysis of Zhuang Yao Shuang Lu Tong Nao Granules in a Rat Model of Ischemic Cerebral Infarction. Anal Cell Pathol (Amst) 2022; 2022:8776079. [PMID: 35846873 PMCID: PMC9277214 DOI: 10.1155/2022/8776079] [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: 04/07/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 12/04/2022] Open
Abstract
This study used a metabolomic approach to reveal changes in the levels of metabolic biomarkers and related metabolic pathways before and after Zhuang Yao Shuang Lu Tong Nao granule (YHT) treatment in rats with cerebral ischemia. The neurological deficit scores were significantly higher in the MCAO_R group than in the NC group, indicating that the mice had significantly impaired motor functions. The YHT group had significantly lower scores than the MCAO_R group, suggesting that YHT significantly improved motor function in rats. TTC staining of the brain tissue revealed that YHT significantly reduced the area of cerebral infarction in the treated rats. The MCAO_R group was better separated from the NC rent, sham, and YHT groups via metabolomic PCA. Moreover, there were significant differences in the differential metabolites between the MACO_R and YHT groups. Eighteen common differential metabolites were detected between the MACO_R and NC groups, MACO_R and sham groups, and MACO_R and YHT groups, indicating that YHT significantly increased the levels of various metabolites in the serum of cerebral ischemic stroke (CIS) rats. Moreover, a total of 23 metabolic pathways were obtained. We identified 11 metabolic pathways with the most significant effects in the bubble plots. In conclusion, from a systems biology perspective, this metabolomics-based study showed that YHT could be used to treat ischemic stroke by modulating changes in endogenous metabolites.
Collapse
|
18
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
19
|
Füzesi MV, Muti IH, Berker Y, Li W, Sun J, Habbel P, Nowak J, Xie Z, Cheng LL, Zhang Y. High Resolution Magic Angle Spinning Proton NMR Study of Alzheimer's Disease with Mouse Models. Metabolites 2022; 12:253. [PMID: 35323696 PMCID: PMC8952313 DOI: 10.3390/metabo12030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is a crippling condition that affects millions of elderly adults each year, yet there remains a serious need for improved methods of diagnosis. Metabolomic analysis has been proposed as a potential methodology to better investigate and understand the progression of this disease; however, studies of human brain tissue metabolomics are challenging, due to sample limitations and ethical considerations. Comprehensive comparisons of imaging measurements in animal models to identify similarities and differences between aging- and AD-associated metabolic changes should thus be tested and validated for future human non-invasive studies. In this paper, we present the results of our highresolution magic angle spinning (HRMAS) nuclear magnetic resonance (NMR) studies of AD and wild-type (WT) mouse models, based on animal age, brain regions, including cortex vs. hippocampus, and disease status. Our findings suggest the ability of HRMAS NMR to differentiate between AD and WT mice using brain metabolomics, which potentially can be implemented in in vivo evaluations.
Collapse
Affiliation(s)
- Mark V. Füzesi
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02115, USA; (M.V.F.); (I.H.M.); (J.S.)
| | - Isabella H. Muti
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02115, USA; (M.V.F.); (I.H.M.); (J.S.)
| | - Yannick Berker
- Hopp Children’s Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany;
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Wei Li
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02115, USA; (W.L.); (Z.X.)
| | - Joseph Sun
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02115, USA; (M.V.F.); (I.H.M.); (J.S.)
| | - Piet Habbel
- Department of Medical Oncology, Haematology and Tumour Immunology, Charité—University Medicine Berlin, 10117 Berlin, Germany;
| | - Johannes Nowak
- Radiology Gotha, SRH Poliklinik Gera, 99867 Gotha, Germany;
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02115, USA; (W.L.); (Z.X.)
| | - Leo L. Cheng
- Departments of Radiology and Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02115, USA; (W.L.); (Z.X.)
| |
Collapse
|
20
|
Discovery of a Metabolic Signature Predisposing High Risk Patients with Mild Cognitive Impairment to Converting to Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222010903. [PMID: 34681563 PMCID: PMC8535253 DOI: 10.3390/ijms222010903] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/17/2022] Open
Abstract
Assessing dementia conversion in patients with mild cognitive impairment (MCI) remains challenging owing to pathological heterogeneity. While many MCI patients ultimately proceed to Alzheimer’s disease (AD), a subset of patients remain stable for various times. Our aim was to characterize the plasma metabolites of nineteen MCI patients proceeding to AD (P-MCI) and twenty-nine stable MCI (S-MCI) patients by untargeted metabolomics profiling. Alterations in the plasma metabolites between the P-MCI and S-MCI groups, as well as between the P-MCI and AD groups, were compared over the observation period. With the help of machine learning-based stratification, a 20-metabolite signature panel was identified that was associated with the presence and progression of AD. Furthermore, when the metabolic signature panel was used for classification of the three patient groups, this gave an accuracy of 73.5% using the panel. Moreover, when specifically classifying the P-MCI and S-MCI subjects, a fivefold cross-validation accuracy of 80.3% was obtained using the random forest model. Importantly, indole-3-propionic acid, a bacteria-generated metabolite from tryptophan, was identified as a predictor of AD progression, suggesting a role for gut microbiota in AD pathophysiology. Our study establishes a metabolite panel to assist in the stratification of MCI patients and to predict conversion to AD.
Collapse
|
21
|
Wang YY, Sun YP, Luo YM, Peng DH, Li X, Yang BY, Wang QH, Kuang HX. Biomarkers for the Clinical Diagnosis of Alzheimer's Disease: Metabolomics Analysis of Brain Tissue and Blood. Front Pharmacol 2021; 12:700587. [PMID: 34366852 PMCID: PMC8333692 DOI: 10.3389/fphar.2021.700587] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/08/2021] [Indexed: 01/09/2023] Open
Abstract
With an increase in aging populations worldwide, age-related diseases such as Alzheimer's disease (AD) have become a global concern. At present, a cure for neurodegenerative disease is lacking. There is an urgent need for a biomarker that can facilitate the diagnosis, classification, prognosis, and treatment response of AD. The recent emergence of highly sensitive mass-spectrometry platforms and high-throughput technology can be employed to discover and catalog vast datasets of small metabolites, which respond to changed status in the body. Metabolomics analysis provides hope for a better understanding of AD as well as the subsequent identification and analysis of metabolites. Here, we review the state-of-the-art emerging candidate biomarkers for AD.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan-Ping Sun
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu-Meng Luo
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Dong-Hui Peng
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiao Li
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bing-You Yang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiu-Hong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hai-Xue Kuang
- Key Laboratory of Chinese Materia Medica Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
22
|
Gallo A, Pillet LE, Verpillot R. New frontiers in Alzheimer's disease diagnostic: Monoamines and their derivatives in biological fluids. Exp Gerontol 2021; 152:111452. [PMID: 34182050 DOI: 10.1016/j.exger.2021.111452] [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: 11/10/2020] [Revised: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
Current diagnosis of Alzheimer's disease (AD) relies on a combination of neuropsychological evaluations, biomarker measurements and brain imaging. Nevertheless, these approaches are either expensive, invasive or lack sensitivity to early AD stages. The main challenge of ongoing research is therefore to identify early non-invasive biomarkers to diagnose AD at preclinical stage. Accumulating evidence support the hypothesis that initial degeneration of profound monoaminergic nuclei may trigger a transneuronal spread of AD pathology towards hippocampus and cortex. These studies aroused great interest on monoamines, i.e. noradrenaline (NA), dopamine (D) ad serotonin (5-HT), as early hallmarks of AD pathology. The present work reviews current literature on the potential role of monoamines and related metabolites as biomarkers of AD. First, morphological changes in the monoaminergic systems during AD are briefly described. Second, we focus on concentration changes of these molecules and their derivatives in biological fluids, including cerebrospinal fluid, obtained by lumbar puncture, and blood or urine, sampled via less invasive procedures. Starting from initial observations, we then discuss recent insights on metabolomics-based analysis, highlighting the promising clinical utility of monoamines for the identification of a molecular AD signature, aimed at improving early diagnosis and discrimination from other dementia.
Collapse
|
23
|
Baumel BS, Doraiswamy PM, Sabbagh M, Wurtman R. Potential Neuroregenerative and Neuroprotective Effects of Uridine/Choline-Enriched Multinutrient Dietary Intervention for Mild Cognitive Impairment: A Narrative Review. Neurol Ther 2021; 10:43-60. [PMID: 33368017 PMCID: PMC8139993 DOI: 10.1007/s40120-020-00227-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/02/2020] [Indexed: 01/21/2023] Open
Abstract
In mild cognitive impairment (MCI) due to Alzheimer disease (AD), also known as prodromal AD, there is evidence for a pathologic shortage of uridine, choline, and docosahexaenoic acid [DHA]), which are key nutrients needed by the brain. Preclinical and clinical evidence shows the importance of nutrient bioavailability to support the development and maintenance of brain structure and function in MCI and AD. Availability of key nutrients is limited in MCI, creating a distinct nutritional need for uridine, choline, and DHA. Evidence suggests that metabolic derangements associated with ageing and disease-related pathology can affect the body's ability to generate and utilize nutrients. This is reflected in lower levels of nutrients measured in the plasma and brains of individuals with MCI and AD dementia, and progressive loss of cognitive performance. The uridine shortage cannot be corrected by normal diet, making uridine a conditionally essential nutrient in affected individuals. It is also challenging to correct the choline shortfall through diet alone, because brain uptake from the plasma significantly decreases with ageing. There is no strong evidence to support the use of single-agent supplements in the management of MCI due to AD. As uridine and choline work synergistically with DHA to increase phosphatidylcholine formation, there is a compelling rationale to combine these nutrients. A multinutrient enriched with uridine, choline, and DHA developed to support brain function has been evaluated in randomized controlled trials covering a spectrum of dementia from MCI to moderate AD. A randomized controlled trial in subjects with prodromal AD showed that multinutrient intervention slowed brain atrophy and improved some measures of cognition. Based on the available clinical evidence, nutritional intervention should be considered as a part of the approach to the management of individuals with MCI due to AD, including adherence to a healthy, balanced diet, and consideration of evidence-based multinutrient supplements.
Collapse
Affiliation(s)
- Barry S Baumel
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Marwan Sabbagh
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Las Vegas, NV, USA
| | - Richard Wurtman
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
24
|
Mechanistic Insights into Alzheimer's Disease Unveiled through the Investigation of Disturbances in Central Metabolites and Metabolic Pathways. Biomedicines 2021; 9:biomedicines9030298. [PMID: 33799385 PMCID: PMC7998757 DOI: 10.3390/biomedicines9030298] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Hydrophilic metabolites are closely involved in multiple primary metabolic pathways and, consequently, play an essential role in the onset and progression of multifactorial human disorders, such as Alzheimer’s disease. This review article provides a comprehensive revision of the literature published on the use of mass spectrometry-based metabolomics platforms for approaching the central metabolome in Alzheimer’s disease research, including direct mass spectrometry, gas chromatography-mass spectrometry, hydrophilic interaction liquid chromatography-mass spectrometry, and capillary electrophoresis-mass spectrometry. Overall, mounting evidence points to profound disturbances that affect a multitude of central metabolic pathways, such as the energy-related metabolism, the urea cycle, the homeostasis of amino acids, fatty acids and nucleotides, neurotransmission, and others.
Collapse
|
25
|
Bustamam MSA, Pantami HA, Azizan A, Shaari K, Min CC, Abas F, Nagao N, Maulidiani M, Banerjee S, Sulaiman F, Ismail IS. Complementary Analytical Platforms of NMR Spectroscopy and LCMS Analysis in the Metabolite Profiling of Isochrysis galbana. Mar Drugs 2021; 19:md19030139. [PMID: 33801258 PMCID: PMC7998644 DOI: 10.3390/md19030139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
This study was designed to profile the metabolites of Isochrysis galbana, an indigenous and less explored microalgae species. 1H Nuclear Magnetic Resonance (NMR) spectroscopy and Liquid Chromatography-Mass Spectrometry (LCMS) were used to establish the metabolite profiles of five different extracts of this microalga, which are hexane (Hex), ethyl acetate (EtOAc), absolute ethanol (EtOH), EtOH:water 1:1 (AqE), and 100% water (Aq). Partial least square discriminant analysis (PLS–DA) of the generated profiles revealed that EtOAc and Aq extracts contain a diverse range of metabolites as compared to the other extracts with a total of twenty-one metabolites, comprising carotenoids, polyunsaturated fatty acids, and amino acids, that were putatively identified from the NMR spectra. Meanwhile, thirty-two metabolites were successfully annotated from the LCMS/MS data, ten of which (palmitic acid, oleic acid, α-linolenic acid, arachidic acid, cholesterol, DHA, DPA, fucoxanthin, astaxanthin, and pheophytin) were similar to those present in the NMR profile. Another eleven glycerophospholipids were discovered using MS/MS-based molecular network (MN) platform. The results of this study, besides providing a better understanding of I.galbana’s chemical make-up, will be of importance in exploring this species potential as a feed ingredient in the aquaculture industry.
Collapse
Affiliation(s)
- Muhammad Safwan Ahamad Bustamam
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.S.A.B.); (A.A.); (K.S.); (F.A.); (S.B.); (F.S.)
| | - Hamza Ahmed Pantami
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Awanis Azizan
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.S.A.B.); (A.A.); (K.S.); (F.A.); (S.B.); (F.S.)
| | - Khozirah Shaari
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.S.A.B.); (A.A.); (K.S.); (F.A.); (S.B.); (F.S.)
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Chong Chou Min
- Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (C.C.M.); (N.N.)
| | - Faridah Abas
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.S.A.B.); (A.A.); (K.S.); (F.A.); (S.B.); (F.S.)
| | - Norio Nagao
- Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (C.C.M.); (N.N.)
| | - Maulidiani Maulidiani
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia;
| | - Sanjoy Banerjee
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.S.A.B.); (A.A.); (K.S.); (F.A.); (S.B.); (F.S.)
| | - Fadzil Sulaiman
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.S.A.B.); (A.A.); (K.S.); (F.A.); (S.B.); (F.S.)
| | - Intan Safinar Ismail
- Natural Medicine and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia; (M.S.A.B.); (A.A.); (K.S.); (F.A.); (S.B.); (F.S.)
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Correspondence: ; Tel.: +60-3-9769-7492
| |
Collapse
|
26
|
de Wit NM, Mol K, Rodríguez-Lorenzo S, de Vries HE, Kooij G. The Role of Sphingolipids and Specialized Pro-Resolving Mediators in Alzheimer's Disease. Front Immunol 2021; 11:620348. [PMID: 33633739 PMCID: PMC7902029 DOI: 10.3389/fimmu.2020.620348] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia worldwide giving rise to devastating forms of cognitive decline, which impacts patients’ lives and that of their proxies. Pathologically, AD is characterized by extracellular amyloid deposition, neurofibrillary tangles and chronic neuroinflammation. To date, there is no cure that prevents progression of AD. In this review, we elaborate on how bioactive lipids, including sphingolipids (SL) and specialized pro-resolving lipid mediators (SPM), affect ongoing neuroinflammatory processes during AD and how we may exploit them for the development of new biomarker panels and/or therapies. In particular, we here describe how SPM and SL metabolism, ranging from ω-3/6 polyunsaturated fatty acids and their metabolites to ceramides and sphingosine-1-phosphate, initiates pro- and anti-inflammatory signaling cascades in the central nervous system (CNS) and what changes occur therein during AD pathology. Finally, we discuss novel therapeutic approaches to resolve chronic neuroinflammation in AD by modulating the SPM and SL pathways.
Collapse
Affiliation(s)
- Nienke M de Wit
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Kevin Mol
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sabela Rodríguez-Lorenzo
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
27
|
Song Z, Wang M, Zhu Z, Tang G, Liu Y, Chai Y. Optimization of pretreatment methods for cerebrospinal fluid metabolomics based on ultrahigh performance liquid chromatography/mass spectrometry. J Pharm Biomed Anal 2021; 197:113938. [PMID: 33621718 DOI: 10.1016/j.jpba.2021.113938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Sample pretreatment of cerebrospinal fluid (CSF) in metabolomics plays an important role in metabolic profiling study, especially for samples related to central nervous system diseases. However, there is few study about optimization of CSF metabolomics pretreatment. Therefore, it is an urgent need to optimize CSF pretreatment in order to promote the extraction efficiency of metabolites. In this study, CSF samples were separately subjected to nine different protein precipitation solvents and five different reconstitution solvents to establish the most effective pretreatment method before hydrophilic interaction (HILIC) and reverse-phase (RP) ultrahigh performance liquid chromatography mass spectrometry (UPLC/MS) analysis. The optimal conditions for different sample pretreatment methods were analyzed based on coverage (number of detected potential metabolites), stability (the relative standard deviation (RSD) distribution of metabolites) and the reproducibility of the data. Our results suggested that using EtOH or MeOH-EtOH-ACN (1:1:1, v/v/v) as the protein precipitation solvents and H2O-MeOH-ACN (2:1:1, v/v/v) as the reconstitution solvent were optimal methods for T3 column analysis. For HILIC column analysis, using EtOH to precipitate protein and H2O-MeOH-ACN (2:1:1, v/v/v) to reconstitute or MeOH to precipitate and 5 %ACN to reconstitute performed best. This developed UPLC/MS pretreatment method could provide better protein precipitation solvents and reconstitution solvents for global CSF metabolic analysis, potentially facilitating the comprehensive understanding of many central nervous system diseases.
Collapse
Affiliation(s)
- Zhiqiang Song
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Mian Wang
- Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Zhenyu Zhu
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China
| | - Gusheng Tang
- Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Yue Liu
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China.
| | - Yifeng Chai
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China.
| |
Collapse
|
28
|
Functional Metabolomics and Chemoproteomics Approaches Reveal Novel Metabolic Targets for Anticancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1280:131-147. [PMID: 33791979 DOI: 10.1007/978-3-030-51652-9_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer cells exhibit different metabolic patterns compared to their normal counterparts. Although the reprogrammed metabolism has been indicated as strong biomarkers of cancer initiation and progression, increasing evidences suggest that metabolic alteration tuned by oncogenic drivers contributes to the occurrence and development of cancers rather than just being a hallmark of cancer. With this notion, targeting cancer metabolism holds promise as a novel anticancer strategy and is embracing its renaissance during the past two decades. Herein we have summarized the most recent developments in omics technology, including both metabolomics and proteomics, and how the combined use of these analytical tools significantly impacts this field by comprehensively and systematically recording the metabolic changes in cancer and hence reveals potential therapeutic targets that function by modulating the disrupted metabolic pathways.
Collapse
|
29
|
Venkataraman L, Fair SR, McElroy CA, Hester ME, Fu H. Modeling neurodegenerative diseases with cerebral organoids and other three-dimensional culture systems: focus on Alzheimer's disease. Stem Cell Rev Rep 2020; 18:696-717. [PMID: 33180261 PMCID: PMC7658915 DOI: 10.1007/s12015-020-10068-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2020] [Indexed: 12/11/2022]
Abstract
Many neurodegenerative diseases (NDs) such as Alzheimer’s disease, Parkinson’s disease, frontotemporal dementia, amyotrophic lateral sclerosis and Huntington’s disease, are characterized by the progressive accumulation of abnormal proteinaceous assemblies in specific cell types and regions of the brain, leading to cellular dysfunction and brain damage. Although animal- and in vitro-based studies of NDs have provided the field with an extensive understanding of some of the mechanisms underlying these diseases, findings from these studies have not yielded substantial progress in identifying treatment options for patient populations. This necessitates the development of complementary model systems that are better suited to recapitulate human-specific features of ND pathogenesis. Three-dimensional (3D) culture systems, such as cerebral organoids generated from human induced pluripotent stem cells, hold significant potential to model NDs in a complex, tissue-like environment. In this review, we discuss the advantages of 3D culture systems and 3D modeling of NDs, especially AD and FTD. We also provide an overview of the challenges and limitations of the current 3D culture systems. Finally, we propose a few potential future directions in applying state-of-the-art technologies in 3D culture systems to understand the mechanisms of NDs and to accelerate drug discovery. Graphical abstract ![]()
Collapse
Affiliation(s)
- Lalitha Venkataraman
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA
| | - Summer R Fair
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH, 43215, USA
- College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Craig A McElroy
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Mark E Hester
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH, 43215, USA.
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University Wexner Medical Center, 616 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
30
|
Wong KY, Roy J, Fung ML, Heng BC, Zhang C, Lim LW. Relationships between Mitochondrial Dysfunction and Neurotransmission Failure in Alzheimer's Disease. Aging Dis 2020; 11:1291-1316. [PMID: 33014538 PMCID: PMC7505271 DOI: 10.14336/ad.2019.1125] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
Besides extracellular deposition of amyloid beta and formation of phosphorylated tau in the brains of patients with Alzheimer's disease (AD), the pathogenesis of AD is also thought to involve mitochondrial dysfunctions and altered neurotransmission systems. However, none of these components can describe the diverse cognitive, behavioural, and psychiatric symptoms of AD without the pathologies interacting with one another. The purpose of this review is to understand the relationships between mitochondrial and neurotransmission dysfunctions in terms of (1) how mitochondrial alterations affect cholinergic and monoaminergic systems via disruption of energy metabolism, oxidative stress, and apoptosis; and (2) how different neurotransmission systems drive mitochondrial dysfunction via increasing amyloid beta internalisation, oxidative stress, disruption of mitochondrial permeabilisation, and mitochondrial trafficking. All these interactions are separately discussed in terms of neurotransmission systems. The association of mitochondrial dysfunctions with alterations in dopamine, norepinephrine, and histamine is the prospective goal in this research field. By unfolding the complex interactions surrounding mitochondrial dysfunction in AD, we can better develop potential treatments to delay, prevent, or cure this devastating disease.
Collapse
Affiliation(s)
- Kan Yin Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Jaydeep Roy
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Man Lung Fung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Boon Chin Heng
- Peking University School of Stomatology, Beijing, China.
| | - Chengfei Zhang
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| | - Lee Wei Lim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
31
|
Neurodegeneration Caused by S1P-Lyase Deficiency Involves Calcium-Dependent Tau Pathology and Abnormal Histone Acetylation. Cells 2020; 9:cells9102189. [PMID: 32998447 PMCID: PMC7599816 DOI: 10.3390/cells9102189] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/03/2023] Open
Abstract
We have shown that sphingosine 1-phosphate (S1P) generated by sphingosine kinase 2 (SK2) is toxic in neurons lacking S1P-lyase (SGPL1), the enzyme that catalyzes its irreversible cleavage. Interestingly, patients harboring mutations in the gene encoding this enzyme (SGPL1) often present with neurological pathologies. Studies in a mouse model with a developmental neural-specific ablation of SGPL1 (SGPL1fl/fl/Nes) confirmed the importance of S1P metabolism for the presynaptic architecture and neuronal autophagy, known to be essential for brain health. We now investigated in SGPL1-deficient murine brains two other factors involved in neurodegenerative processes, namely tau phosphorylation and histone acetylation. In hippocampal and cortical slices SGPL1 deficiency and hence S1P accumulation are accompanied by hyperphosphorylation of tau and an elevated acetylation of histone3 (H3) and histone4 (H4). Calcium chelation with BAPTA-AM rescued both tau hyperphosphorylation and histone acetylation, designating calcium as an essential mediator of these (patho)physiological functions of S1P in the brain. Studies in primary cultured neurons and astrocytes derived from SGPL1fl/fl/Nes mice revealed hyperphosphorylated tau only in SGPL1-deficient neurons and increased histone acetylation only in SGPL1-deficient astrocytes. Both could be reversed to control values with BAPTA-AM, indicating the close interdependence of S1P metabolism, calcium homeostasis, and brain health.
Collapse
|
32
|
Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer's Disease: Sensitivity, Specificity and Potential for Clinical Use. J Pers Med 2020; 10:jpm10030116. [PMID: 32911755 PMCID: PMC7565390 DOI: 10.3390/jpm10030116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, affecting more than 5 million Americans, with steadily increasing mortality and incredible socio-economic burden. Not only have therapeutic efforts so far failed to reach significant efficacy, but the real pathogenesis of the disease is still obscure. The current theories are based on pathological findings of amyloid plaques and tau neurofibrillary tangles that accumulate in the brain parenchyma of affected patients. These findings have defined, together with the extensive neurodegeneration, the diagnostic criteria of the disease. The ability to detect changes in the levels of amyloid and tau in cerebrospinal fluid (CSF) first, and more recently in blood, has allowed us to use these biomarkers for the specific in-vivo diagnosis of AD in humans. Furthermore, other pathological elements of AD, such as the loss of neurons, inflammation and metabolic derangement, have translated to the definition of other CSF and blood biomarkers, which are not specific of the disease but, when combined with amyloid and tau, correlate with the progression from mild cognitive impairment to AD dementia, or identify patients who will develop AD pathology. In this review, we discuss the role of current and hypothetical biomarkers of Alzheimer's disease, their specificity, and the caveats of current high-sensitivity platforms for their peripheral detection.
Collapse
|
33
|
Hajjar I, Liu C, Jones DP, Uppal K. Untargeted metabolomics reveal dysregulations in sugar, methionine, and tyrosine pathways in the prodromal state of AD. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12064. [PMID: 32793799 PMCID: PMC7418891 DOI: 10.1002/dad2.12064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Altered metabolism may occur years before clinical manifestations of Alzheimer's disease (AD). We used untargeted metabolomics on the cerebrospinal fluid of patients with mild cognitive impairment (MCI) to uncover metabolomic derangements. METHODS CSF from 92 normal controls and 93 MCI underwent untargeted metabolomics using high-resolution mass spectrometry with liquid chromatography. Partial least squares discriminant analysis was used followed by metabolite annotation and pathway enrichment analysis (PES). Significant features were correlated with disease phenotypes. RESULTS We identified 294 features differentially expressed between the two groups and 94 were annotated. PES showed that sugar regulation (N-glycan, P = .0007; sialic acid, P = .0014; aminosugars, P = .0042; galactose, P = .0054), methionine regulation (P = .0081), and tyrosine metabolism (P = .019) pathways were differentially activated and significant features within these pathways correlated with multiple disease phenotypes. CONCLUSION There is a metabolic signature characterized by impairments in sugars, methionine, and tyrosine regulation in MCI. Targeting these pathways may offer new therapeutic approaches to AD.
Collapse
Affiliation(s)
- Ihab Hajjar
- Medicine and NeurologyDepartment of NeurologyEmory UniversityAtlantaGeorgiaUSA
| | - Chang Liu
- Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Dean P. Jones
- Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Karan Uppal
- Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
34
|
Sengupta A, Weljie AM. Metabolism of sleep and aging: Bridging the gap using metabolomics. NUTRITION AND HEALTHY AGING 2019; 5:167-184. [PMID: 31984245 PMCID: PMC6971829 DOI: 10.3233/nha-180043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sleep is a conserved behavior across the evolutionary timescale. Almost all known animal species demonstrate sleep or sleep like states. Despite extensive study, the mechanistic aspects of sleep need are not very well characterized. Sleep appears to be needed to generate resources that are utilized during the active stage/wakefulness as well as clearance of waste products that accumulate during wakefulness. From a metabolic perspective, this means sleep is crucial for anabolic activities. Decrease in anabolism and build-up of harmful catabolic waste products is also a hallmark of aging processes. Through this lens, sleep and aging processes are remarkably parallel- for example behavioral studies demonstrate an interaction between sleep and aging. Changes in sleep behavior affect neurocognitive phenotypes important in aging such as learning and memory, although the underlying connections are largely unknown. Here we draw inspiration from the similar metabolic effects of sleep and aging and posit that large scale metabolic phenotyping, commonly known as metabolomics, can shed light to interleaving effects of sleep, aging and progression of diseases related to aging. In this review, data from recent sleep and aging literature using metabolomics as principal molecular phenotyping methods is collated and compared. The present data suggests that metabolic effects of aging and sleep also demonstrate similarities, particularly in lipid metabolism and amino acid metabolism. Some of these changes also overlap with metabolomic data available from clinical studies of Alzheimer's disease. Together, metabolomic technologies show promise in elucidating interleaving effects of sleep, aging and progression of aging disorders at a molecular level.
Collapse
Affiliation(s)
- Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Aalim M. Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
35
|
Niedzwiecki MM, Walker DI, Howell JC, Watts KD, Jones DP, Miller GW, Hu WT. High-resolution metabolomic profiling of Alzheimer's disease in plasma. Ann Clin Transl Neurol 2019; 7:36-45. [PMID: 31828981 PMCID: PMC6952314 DOI: 10.1002/acn3.50956] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 12/13/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a complex neurological disorder with contributions from genetic and environmental factors. High‐resolution metabolomics (HRM) has the potential to identify novel endogenous and environmental factors involved in AD. Previous metabolomics studies have identified circulating metabolites linked to AD, but lack of replication and inconsistent diagnostic algorithms have hindered the generalizability of these findings. Here we applied HRM to identify plasma metabolic and environmental factors associated with AD in two study samples, with cerebrospinal fluid (CSF) biomarkers of AD incorporated to achieve high diagnostic accuracy. Methods Liquid chromatography‐mass spectrometry (LC–MS)‐based HRM was used to identify plasma and CSF metabolites associated with AD diagnosis and CSF AD biomarkers in two studies of prevalent AD (Study 1: 43 AD cases, 45 mild cognitive impairment [MCI] cases, 41 controls; Study 2: 50 AD cases, 18 controls). AD‐associated metabolites were identified using a metabolome‐wide association study (MWAS) framework. Results An MWAS meta‐analysis identified three non‐medication AD‐associated metabolites in plasma, including elevated levels of glutamine and an unknown halogenated compound and lower levels of piperine, a dietary alkaloid. The non‐medication metabolites were correlated with CSF AD biomarkers, and glutamine and the unknown halogenated compound were also detected in CSF. Furthermore, in Study 1, the unknown compound and piperine were altered in MCI patients in the same direction as AD dementia. Conclusions In plasma, AD was reproducibly associated with elevated levels of glutamine and a halogen‐containing compound and reduced levels of piperine. These findings provide further evidence that exposures and behavior may modify AD risks.
Collapse
Affiliation(s)
- Megan M Niedzwiecki
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Douglas I Walker
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York.,Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia
| | | | - Kelly D Watts
- Department of Neurology, Emory University, Atlanta, Georgia
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia
| | - Gary W Miller
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Department of Neurology, Emory University, Atlanta, Georgia.,Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia.,Department of Pharmacology, Emory University, Atlanta, Georgia
| | - William T Hu
- Department of Neurology, Emory University, Atlanta, Georgia.,Center for Neurodegenerative Diseases, Emory University, Atlanta, Georgia.,Alzheimer's Disease Research Center, Emory University, Atlanta, Georgia
| |
Collapse
|
36
|
Costa AC, Joaquim HPG, Forlenza O, Talib LL, Gattaz WF. Plasma lipids metabolism in mild cognitive impairment and Alzheimer's disease. World J Biol Psychiatry 2019; 20:190-196. [PMID: 28922966 DOI: 10.1080/15622975.2017.1369566] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Expression of phospholipids and related molecules could provide panels of multiple biomarkers searching for the signature of Alzheimer's disease (AD). The aim of the present study was to quantify ten phospholipids and simultaneously determine phospholipase A2 (PLA2) activity in blood of mild cognitive impairment (MCI) and AD patients. METHODS Thirty-four AD, 20 MCI and 25 controls were enrolled. The phospholipids where analysed using the AbsoluteIDQ® p180 Kit. PLA2 activities were accessed in platelets by a radio-enzymatic assay. RESULTS The study failed to fix the ten phospholipids as a panel to predict AD; the levels of PCaaC36:6, PCaaC40:6 and C16:1-OH were lower in MCI than in controls (P = 0.041, P = 0.012, P = 0.044 respectively). PCaaC40:2 levels were lower in MCI than in AD (P = 0.041). The converters MCI-AD showed at baseline lower levels of PCaaC40:2 (P = 0.050) and PCaaC40:6 (P = 0.037) than controls. iPLA2 activity was reduced in AD and MCI than in controls (P < 0.001). We found positive correlation in the control group between PCaaC38:6 and tPLA2 (r = 0.680; P = 0.001) and sPLA2 (r = 0.601; P = 0.004); PCaaC40:1 and iPLA2 (r = 0.503; P = 0.020); PCaaC40:6 and tPLA2 (r = 0.532; P = 0.013) and sPLA2 (r = 0.523; P = 0.015). CONCLUSIONS Lipids metabolites in plasma might indirectly indicate changes in neuronal membrane and this deregulation can outline the transition between healthy and diseased brains.
Collapse
Affiliation(s)
- Alana C Costa
- a Department and Institute of Psychiatry , Laboratory of Neurosciences (LIM 27), University of São Paulo (USP) , São Paulo , Brazil
| | - Helena P G Joaquim
- a Department and Institute of Psychiatry , Laboratory of Neurosciences (LIM 27), University of São Paulo (USP) , São Paulo , Brazil
| | - Orestes Forlenza
- a Department and Institute of Psychiatry , Laboratory of Neurosciences (LIM 27), University of São Paulo (USP) , São Paulo , Brazil
| | - Leda L Talib
- a Department and Institute of Psychiatry , Laboratory of Neurosciences (LIM 27), University of São Paulo (USP) , São Paulo , Brazil
| | - Wagner F Gattaz
- a Department and Institute of Psychiatry , Laboratory of Neurosciences (LIM 27), University of São Paulo (USP) , São Paulo , Brazil
| |
Collapse
|
37
|
Zhang M, Liu Y, Liu M, Liu B, Li N, Dong X, Hong Z, Chai Y. UHPLC-QTOF/MS-based metabolomics investigation for the protective mechanism of Danshen in Alzheimer's disease cell model induced by Aβ 1-42. Metabolomics 2019; 15:13. [PMID: 30830431 DOI: 10.1007/s11306-019-1473-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a chronic neurodegenerative disorder with neither definitive pathogenesis nor effective therapy so far. Danshen, the dried root and rhizome of Salvia miltiorrhiza Bunge, is used extensively in Alzheimer's disease treatment to ameliorate the symptoms, but the underlying mechanism remains to be clarified. OBJECTIVES To investigate potential biomarkers for AD and elucidate the protective mechanism of Danshen on AD cell model. METHODS An ultra high performance liquid chromatography-quadrupole time of flight mass spectrometry (UHPLC-QTOF/MS)-based approach combined with partial least squares discriminant analysis (PLS-DA) has been developed to discriminate the metabolic modifications between human brain microvascular endothelial cell (hBMEC) and AD cell model induced by amyloid-β protein (Aβ1-42). To further elucidate the pathophysiology of AD, related metabolic pathways have been studied. RESULTS Thirty-three distinct potential biomarkers were screened out and considered as potential biomarkers corresponding to AD, which were mostly improved and partially restored back to normalcy in Danshen pre-protection group. It was found that AD was closely related to disturbed arginine and proline metabolism, glutathione metabolism, alanine aspartate and glutamate metabolism, histidine metabolism, pantothenate and CoA biosynthesis, phenylalanine tyrosine and tryptophan biosynthesis, citrate cycle and glycerophospholipid metabolism, and the protective mechanism of Danshen in AD cell model may be related to partially regulating the perturbed pathways. CONCLUSIONS These outcomes provide valuable evidences for therapeutic mechanism investigation of Danshen in AD treatment, and such an approach could be transferred to unravel the mechanism of other traditional Chinese medicine (TCM) and diseases.
Collapse
Affiliation(s)
- Mingyong Zhang
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China
| | - Yue Liu
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China
| | - Min Liu
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Biying Liu
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Na Li
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Xin Dong
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Zhanying Hong
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China.
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China.
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| |
Collapse
|
38
|
Zhong L, Jiang X, Zhu Z, Qin H, Dinkins MB, Kong JN, Leanhart S, Wang R, Elsherbini A, Bieberich E, Zhao Y, Wang G. Lipid transporter Spns2 promotes microglia pro-inflammatory activation in response to amyloid-beta peptide. Glia 2018; 67:498-511. [PMID: 30484906 DOI: 10.1002/glia.23558] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/31/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022]
Abstract
Accumulating evidence indicates that neuroinflammation contributes to the pathogenesis and exacerbation of neurodegenerative disorders, such as Alzheimer's disease (AD). Sphingosine-1-phosphate (S1P) is a pleiotropic bioactive lipid that regulates many pathophysiological processes including inflammation. We present evidence here that the spinster homolog 2 (Spns2), a S1P transporter, promotes microglia pro-inflammatory activation in vitro and in vivo. Spns2 knockout (Spns2KO) in primary cultured microglia resulted in significantly reduced levels of pro-inflammatory cytokines induced by lipopolysaccharide (LPS) and amyloid-beta peptide 1-42 oligomers (Aβ42) when compared with littermate controls. Fingolimod (FTY720), a S1P receptor 1 (S1PR1) functional antagonist and FDA approved drug for relapsing-remitting multiple sclerosis, partially blunted Aβ42-induced pro-inflammatory cytokine generation, suggesting that Spns2 promotes microglia pro-inflammatory activation through S1P-signaling. Spns2KO significantly reduced Aβ42-induced nuclear factor kappa B (NFκB) activity. S1P increased, while FTY720 dampened, Aβ42-induced NFκB activity, suggesting that Spns2 activates microglia inflammation through, at least partially, NFκB pathway. Spns2KO mouse brains showed significantly reduced Aβ42-induced microglia activation/accumulation and reduced levels of pro-inflammatory cytokines when compared with age-matched controls. More interestingly, Spns2KO ameliorated Aβ42-induced working memory deficit detected by Y-Maze. In summary, these results suggest that Spns2 promotes pro-inflammatory polarization of microglia and may play a crucial role in AD pathogenesis.
Collapse
Affiliation(s)
- Liansheng Zhong
- Department of Physiology, University of Kentucky, Lexington, Kentucky.,Department of Bioinformatics, Key Laboratory of Cell Biology of Ministry of Public Health, College of Life Sciences, China Medical University, Shenyang, China
| | - Xue Jiang
- Department of Physiology, University of Kentucky, Lexington, Kentucky.,Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - Zhihui Zhu
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Haiyan Qin
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Michael B Dinkins
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Ji-Na Kong
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Silvia Leanhart
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Rebecca Wang
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Ahmed Elsherbini
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky, Lexington, Kentucky.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yujie Zhao
- Department of Bioinformatics, Key Laboratory of Cell Biology of Ministry of Public Health, College of Life Sciences, China Medical University, Shenyang, China
| | - Guanghu Wang
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
39
|
Sapkota S, Huan T, Tran T, Zheng J, Camicioli R, Li L, Dixon RA. Alzheimer's Biomarkers From Multiple Modalities Selectively Discriminate Clinical Status: Relative Importance of Salivary Metabolomics Panels, Genetic, Lifestyle, Cognitive, Functional Health and Demographic Risk Markers. Front Aging Neurosci 2018; 10:296. [PMID: 30333744 PMCID: PMC6175993 DOI: 10.3389/fnagi.2018.00296] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Among the neurodegenerative diseases of aging, sporadic Alzheimer’s disease (AD) is the most prevalent and perhaps the most feared. With virtually no success at finding pharmaceutical therapeutics for altering progressive AD after diagnosis, research attention is increasingly directed at discovering biological and other markers that detect AD risk in the long asymptomatic phase. Both early detection and precision preclinical intervention require systematic investigation of multiple modalities and combinations of AD-related biomarkers and risk factors. We extend recent unbiased metabolomics research that produced a set of metabolite biomarker panels tailored to the discrimination of cognitively normal (CN), cognitively impaired and AD patients. Specifically, we compare the prediction importance of these panels with five other sets of modifiable and non-modifiable AD risk factors (genetic, lifestyle, cognitive, functional health and bio-demographic) in three clinical groups. Method: The three groups were: CN (n = 35), mild cognitive impairment (MCI; n = 25), and AD (n = 22). In a series of three pairwise comparisons, we used machine learning technology random forest analysis (RFA) to test relative predictive importance of up to 19 risk biomarkers from the six AD risk domains. Results: The three RFA multimodal prediction analyses produced significant discriminating risk factors. First, discriminating AD from CN was the AD metabolite panel and two cognitive markers. Second, discriminating AD from MCI was the AD/MCI metabolite panel and two cognitive markers. Third, discriminating MCI from CN was the MCI metabolite panel and seven markers from four other risk modalities: genetic, lifestyle, cognition and functional health. Conclusions: Salivary metabolomics biomarker panels, supplemented by other risk markers, were robust predictors of: (1) clinical differences in impairment and dementia and even; (2) subtle differences between CN and MCI. For the latter, the metabolite panel was supplemented by biomarkers that were both modifiable (e.g., functional) and non-modifiable (e.g., genetic). Comparing, integrating and identifying important multi-modal predictors may lead to novel combinations of complex risk profiles potentially indicative of neuropathological changes in asymptomatic or preclinical AD.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Tao Huan
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Tran Tran
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jiamin Zheng
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.,Department of Medicine (Neurology), University of Alberta, Edmonton, AB, Canada
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Roger A Dixon
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.,Department of Psychology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
40
|
Next-generation biomarker discovery in Alzheimer's disease using metabolomics - from animal to human studies. Bioanalysis 2018; 10:1525-1546. [PMID: 30198770 DOI: 10.4155/bio-2018-0135] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a complex disease driven mainly by neuronal loss due to accumulation of intracellular neurofibrillary tangles and amyloid β aggregates in the brain. The diagnosis of AD currently relies on clinical symptoms while the disease can only be confirmed at autopsy. The few available biomarkers allowing for diagnosis are typically detected many years after the onset of the disease. New diagnostic approaches, particularly in easily-accessible biofluids, are essential. By providing an exhaustive information of the phenotype, metabolomics is an ideal approach for identification of new biomarkers. This review investigates the current position of metabolomics in the field of AD research, focusing on animal and human studies, and discusses the improvements carried out over the past decade.
Collapse
|
41
|
Advances and challenges in neurochemical profiling of biological samples using mass spectrometry coupled with separation methods. Trends Analyt Chem 2018. [DOI: 10.1016/j.trac.2018.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
42
|
Hao L, Wang J, Page D, Asthana S, Zetterberg H, Carlsson C, Okonkwo OC, Li L. Comparative Evaluation of MS-based Metabolomics Software and Its Application to Preclinical Alzheimer's Disease. Sci Rep 2018; 8:9291. [PMID: 29915347 PMCID: PMC6006240 DOI: 10.1038/s41598-018-27031-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/17/2018] [Indexed: 02/07/2023] Open
Abstract
Mass spectrometry-based metabolomics has undergone significant progresses in the past decade, with a variety of software packages being developed for data analysis. However, systematic comparison of different metabolomics software tools has rarely been conducted. In this study, several representative software packages were comparatively evaluated throughout the entire pipeline of metabolomics data analysis, including data processing, statistical analysis, feature selection, metabolite identification, pathway analysis, and classification model construction. LC-MS-based metabolomics was applied to preclinical Alzheimer's disease (AD) using a small cohort of human cerebrospinal fluid (CSF) samples (N = 30). All three software packages, XCMS Online, SIEVE, and Compound Discoverer, provided consistent and reproducible data processing results. A hybrid method combining statistical test and support vector machine feature selection was employed to screen key metabolites, achieving a complementary selection of candidate biomarkers from three software packages. Machine learning classification using candidate biomarkers generated highly accurate and predictive models to classify patients into preclinical AD or control category. Overall, our study demonstrated a systematic evaluation of different MS-based metabolomics software packages for the entire data analysis pipeline which was applied to the candidate biomarker discovery of preclinical AD.
Collapse
Affiliation(s)
- Ling Hao
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | | | - David Page
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital Mölndal, Mölndal, Sweden.,Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.,Dementia Research Institute, London, UK
| | - Cynthia Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA. .,Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
43
|
Nagata Y, Hirayama A, Ikeda S, Shirahata A, Shoji F, Maruyama M, Kayano M, Bundo M, Hattori K, Yoshida S, Goto YI, Urakami K, Soga T, Ozaki K, Niida S. Comparative analysis of cerebrospinal fluid metabolites in Alzheimer's disease and idiopathic normal pressure hydrocephalus in a Japanese cohort. Biomark Res 2018; 6:5. [PMID: 29387418 PMCID: PMC5778653 DOI: 10.1186/s40364-018-0119-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/03/2018] [Indexed: 01/02/2023] Open
Abstract
Background Alzheimer’s disease (AD) is a most common dementia in elderly people. Since AD symptoms resemble those of other neurodegenerative diseases, including idiopathic normal pressure hydrocephalus (iNPH), it is difficult to distinguish AD from iNPH for a precise and early diagnosis. iNPH is caused by the accumulation of cerebrospinal fluid (CSF) and involves gait disturbance, urinary incontinence, and dementia. iNPH is treatable with shunt operation which removes accumulated CSF from the brain ventricles. Methods We performed metabolomic analysis in the CSF of patients with AD and iNPH with capillary electrophoresis-mass spectrometry. We assessed metabolites to discriminate between AD and iNPH with Welch’s t-test, receiver operating characteristic (ROC) curve analysis, and multiple logistic regression analysis. Results We found significant increased levels of glycerate and N-acetylneuraminate and significant decreased levels of serine and 2-hydroxybutyrate in the CSF of patients with AD compared to the CSF of patients with iNPH. The ROC curve analysis with these four metabolites showed that the area under the ROC curve was 0.90, indicating good discrimination between AD and iNPH. Conclusions This study identified four metabolites that could possibly discriminate between AD and iNPH, which previous research has shown are closely related to the risk factors, pathogenesis, and symptoms of AD. Analyzing pathway-specific metabolites in the CSF of patients with AD may further elucidate the mechanism and pathogenesis of AD. Electronic supplementary material The online version of this article (10.1186/s40364-018-0119-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuki Nagata
- 1Medical Genome Center, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan
| | - Akiyoshi Hirayama
- 2Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052 Japan
| | - Satsuki Ikeda
- 2Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052 Japan
| | - Aoi Shirahata
- 2Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052 Japan
| | - Futaba Shoji
- 2Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052 Japan
| | - Midori Maruyama
- 2Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052 Japan
| | - Mitsunori Kayano
- 3Research Center for Global Agromedicine, Obihiro University of Agriculture and Veterinary Medicine, 2-11 Inada-cho, Obihiro, Hokkaido 080-8555 Japan
| | - Masahiko Bundo
- 4Department of Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511 Japan
| | - Kotaro Hattori
- 5Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8551 Japan
| | - Sumiko Yoshida
- 5Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8551 Japan
| | - Yu-Ichi Goto
- 5Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8551 Japan
| | - Katsuya Urakami
- 6Department of Biological Regulation, School of Health Science, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503 Japan
| | - Tomoyoshi Soga
- 2Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052 Japan
| | - Kouichi Ozaki
- 1Medical Genome Center, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan
| | - Shumpei Niida
- 1Medical Genome Center, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan
| |
Collapse
|
44
|
Forgacsova A, Galba J, Garruto RM, Majerova P, Katina S, Kovac A. A novel liquid chromatography/mass spectrometry method for determination of neurotransmitters in brain tissue: Application to human tauopathies. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1073:154-162. [PMID: 29275172 DOI: 10.1016/j.jchromb.2017.12.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/06/2017] [Accepted: 12/08/2017] [Indexed: 12/29/2022]
Abstract
Neurotransmitters, small molecules widely distributed in the central nervous system are essential in transmitting electrical signals across neurons via chemical communication. Dysregulation of these chemical signaling molecules is linked to numerous neurological diseases including tauopathies. In this study, a precise and reliable liquid chromatography method was established with tandem mass spectrometry detection for the simultaneous determination of aspartic acid, asparagine, glutamic acid, glutamine, γ-aminobutyric acid, N-acetyl-l-aspartic acid, pyroglutamic acid, acetylcholine and choline in human brain tissue. The method was successfully applied to the analysis of human brain tissues from three different tauopathies; corticobasal degeneration, progressive supranuclear palsy and parkinsonism-dementia complex of Guam. Neurotransmitters were analyzed on ultra-high performance chromatography (UHPLC) using an ethylene bridged hybrid amide column coupled with tandem mass spectrometry (MS/MS). Identification and quantification of neurotransmitters was carried out by ESI+ mass spectrometry detection. We optimized sample preparation to achieve simple and fast extraction of all nine analytes. Our method exhibited an excellent linearity for all analytes (all coefficients of determination >0.99), with inter-day and intra-day precision yielding relative standard deviations 3.2%-11.2% and an accuracy was in range of 92.6%-104.3%. The present study, using the above method, is the first to demonstrate significant alterations of brain neurotransmitters caused by pathological processes in the brain tissues of patient with three different tauopathies.
Collapse
Affiliation(s)
- Andrea Forgacsova
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy of Comenius University, Odbojarov 10, 832 32, Bratislava, Slovak Republic.
| | - Jaroslav Galba
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy of Comenius University, Odbojarov 10, 832 32, Bratislava, Slovak Republic; AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovak Republic
| | - Ralph M Garruto
- Graduate Program in Biomedical Anthropology, Departments of Anthropology and Biological Sciences, Binghamton University, Binghamton, NY, USA; Institute of Mathematics and Statistics, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37, Brno, Czech Republic
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510, Bratislava, Slovak Republic; AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovak Republic
| | - Stanislav Katina
- Institute of Mathematics and Statistics, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37, Brno, Czech Republic
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 84510, Bratislava, Slovak Republic; Department of Pharmacology and Toxicology, The University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181, Kosice, Slovak Republic; AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovak Republic
| |
Collapse
|
45
|
González-Domínguez R, Sayago A, Fernández-Recamales Á. Metabolomics in Alzheimer’s disease: The need of complementary analytical platforms for the identification of biomarkers to unravel the underlying pathology. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1071:75-92. [DOI: 10.1016/j.jchromb.2017.02.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 01/27/2017] [Accepted: 02/05/2017] [Indexed: 12/14/2022]
|
46
|
Yi L, Liu W, Wang Z, Ren D, Peng W. Characterizing Alzheimer's disease through metabolomics and investigating anti-Alzheimer's disease effects of natural products. Ann N Y Acad Sci 2017. [PMID: 28632966 DOI: 10.1111/nyas.13385] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Lunzhao Yi
- Yunnan Food Safety Research Institute; Kunming University of Science and Technology; Kunming China
| | - Wenbin Liu
- Yunnan Food Safety Research Institute; Kunming University of Science and Technology; Kunming China
| | - Zhe Wang
- Department of Integrated Traditional Chinese & Western Medicine, the Second Xiangya Hospital; Central South University; Changsha Hunan China
| | - Dabing Ren
- Yunnan Food Safety Research Institute; Kunming University of Science and Technology; Kunming China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, the Second Xiangya Hospital; Central South University; Changsha Hunan China
| |
Collapse
|
47
|
van Wijk N, Slot RER, Duits FH, Strik M, Biesheuvel E, Sijben JWC, Blankenstein MA, Bierau J, van der Flier WM, Scheltens P, Teunissen CE. Nutrients required for phospholipid synthesis are lower in blood and cerebrospinal fluid in mild cognitive impairment and Alzheimer's disease dementia. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2017; 8:139-146. [PMID: 28653034 PMCID: PMC5476966 DOI: 10.1016/j.dadm.2017.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Synaptic membrane formation depends on nutrients that fuel metabolic pathways for the synthesis of constituent phospholipids. Consequently, insufficient availability of such nutrients may restrict membrane formation and contribute to synaptic dysfunction in Alzheimer's disease (AD). We assessed whether blood and cerebrospinal fluid (CSF) concentrations of nutrients related to phospholipid synthesis differ among patients with AD, mild cognitive impairment (MCI), and control subjects. METHODS Concentrations of uridine, choline, folate, homocysteine, and other related metabolites were analyzed in paired blood and CSF samples from subjects selected from the Amsterdam Dementia Cohort with AD (n = 150; age, 66 ± 7 years; 37% female), MCI (n = 148; age, 66 ± 8 years; 37% female), and control subjects (n = 148; age, 59 ± 8 years; 38% female). RESULTS Age- and gender-adjusted analysis of variance revealed different concentrations of circulating uridine, choline, and folate and CSF uridine, folate, and homocysteine (all P < .05) among the three diagnostic groups. Post hoc pairwise comparison showed that subjects with AD had lower CSF uridine, plasma choline and higher CSF homocysteine concentrations, whereas subjects with MCI had lower plasma and CSF uridine, serum and CSF folate, and higher CSF homocysteine concentrations compared with control subjects (all P < .05), with differences ranging from -11 to +22%. DISCUSSION AD and MCI patients have lower levels of nutrients involved in phospholipid synthesis. The current observations warrant exploration of the application of nutritional strategies in the early stages of AD.
Collapse
Affiliation(s)
- Nick van Wijk
- Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Rosalinde E R Slot
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Flora H Duits
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Marieke Strik
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Egbert Biesheuvel
- Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - John W C Sijben
- Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Marinus A Blankenstein
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Jörgen Bierau
- Department of Clinical Genetics, Maastricht UMC+, Maastricht, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Enche Ady CNA, Lim SM, Teh LK, Salleh MZ, Chin AV, Tan MP, Poi PJH, Kamaruzzaman SB, Abdul Majeed AB, Ramasamy K. Metabolomic-guided discovery of Alzheimer's disease biomarkers from body fluid. J Neurosci Res 2017; 95:2005-2024. [PMID: 28301062 DOI: 10.1002/jnr.24048] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/31/2017] [Accepted: 02/15/2017] [Indexed: 12/11/2022]
Abstract
The rapid increase in the older population has made age-related diseases like Alzheimer's disease (AD) a global concern. Given that there is still no cure for this neurodegenerative disease, the drastic growth in the number of susceptible individuals represents a major emerging threat to public health. The poor understanding of the mechanisms underlying AD is deemed the greatest stumbling block against progress in definitive diagnosis and management of this disease. There is a dire need for biomarkers that can facilitate early diagnosis, classification, prognosis, and treatment response. Efforts have been directed toward discovery of reliable and distinctive AD biomarkers but with very little success. With the recent emergence of high-throughput technology that is able to collect and catalogue vast datasets of small metabolites, metabolomics offers hope for a better understanding of AD and subsequent identification of biomarkers. This review article highlights the potential of using multiple metabolomics platforms as useful means in uncovering AD biomarkers from body fluids. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Che Nor Adlia Enche Ady
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Lay Kek Teh
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Mohd Zaki Salleh
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Ai-Vyrn Chin
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Maw Pin Tan
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Philip Jun Hua Poi
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Shahrul Bahyah Kamaruzzaman
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Abu Bakar Abdul Majeed
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Brain Degeneration and Therapeutics Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Kalavathy Ramasamy
- Faculty of Pharmacy, University Teknologi MARA (UiTM), 42300 Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical and Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
49
|
Li H, Wang M, Liang Q, Jin S, Sun X, Jiang Y, Pan X, Zhou Y, Peng Y, Zhang B, Zhou A, Zhang Y, Chen Z, Cao J, Zhang H, Xia W, Zheng T, Cai Z, Li Y, Xu S. Urinary metabolomics revealed arsenic exposure related to metabolic alterations in general Chinese pregnant women. J Chromatogr A 2017; 1479:145-152. [DOI: 10.1016/j.chroma.2016.12.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/16/2016] [Accepted: 12/03/2016] [Indexed: 10/20/2022]
|
50
|
Khan F, Oloketuyi SF. A future perspective on neurodegenerative diseases: nasopharyngeal and gut microbiota. J Appl Microbiol 2016; 122:306-320. [PMID: 27740729 DOI: 10.1111/jam.13327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/23/2016] [Accepted: 10/08/2016] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases are considered a serious life-threatening issue regardless of age. Resulting nerve damage progressively affects important activities, such as movement, coordination, balance, breathing, speech and the functioning of vital organs. Reports on the subject have concluded that neurodegenerative disease can be caused by mutations of susceptible genes, alcohol consumption, toxins, chemicals and other unknown environmental factors. Although several diagnostic techniques can be used to determine aetiologies, the process is difficult and often fails. Research shows that nasopharyngeal and gut microbiota play important roles in brain to spinal cord coordination. However, no conclusive epidemiologic evidence is available on the roles played by respiratory and gut microbiota in the development of neurodegenerative diseases. Thus, understanding the connection between respiratory and gut microbiota and the nervous system could provide information on causal links. The present review describes future perspectives on the role played by nasopharyngeal and gut microbiota in the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- F Khan
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, UP, India
| | - S F Oloketuyi
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, UP, India
| |
Collapse
|