1
|
Martínez-Gardeazabal J, Pereira-Castelo G, Moreno-Rodríguez M, Llorente-Ovejero A, Fernández M, Fernández-Vega I, Manuel I, Rodríguez-Puertas R. Sphingosine 1-phosphate receptor subtype 1 (S1P 1) activity in the course of Alzheimer's disease. Neurobiol Dis 2024; 202:106713. [PMID: 39448041 DOI: 10.1016/j.nbd.2024.106713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024] Open
Abstract
Some specific lipid molecules in the brain act as signaling molecules, neurotransmitters, or neuromodulators, by binding to specific G protein-coupled receptors (GPCR) for neurolipids. One such receptor, sphingosine 1-phosphate receptor subtype 1 (S1P1), is coupled to Gi/o proteins and is involved in cell proliferation, growth, and neuroprotection. S1P1 constitutes an interesting target for neurodegenerative diseases like multiple sclerosis and Alzheimer's disease (AD), in which changes in the sphingolipid metabolism have been observed. This study analyzes S1P1 receptor-mediated activity in healthy brains and during AD progression using postmortem samples from controls and patients at different Braak's stages. Additionally, the distribution of S1P1 receptor activity in human brains is compared to that in commonly used rodent models, rats and mice, through functional autoradiography, measuring [35S]GTPγS binding stimulated by the S1P1 receptor selective agonist CYM-5442 to obtain the distribution of functional activity of S1P1 receptors. S1P1 receptor-mediated activity, along with that of the cannabinoid CB1 receptor, is one of the highest recorded for any GPCR in many gray matter areas of the brain, reaching maximum values in the cerebellar cortex, specific areas of the hippocampus and the basal forebrain. S1P1 signaling is crucial in areas that regulate learning, memory, motor control, and nociception, such as the basal forebrain and basal ganglia. In AD, S1P1 receptor activity is increased in the inner layers of the frontal cortex and underlying cortical white matter at early stages, but decreases in the hippocampus in advanced stages, indicating ongoing brain impairment. Importantly, we identified significant correlations between S1P1 receptor activity and Braak stages, suggesting that S1P1 receptor dysfunction is associated to disease progression, particularly in memory-related regions. The S1P signaling via S1P1 receptor is a promising neurological target due to its role in key neurophysiological functions and its potential to modify the progression of neurodegenerative diseases. Finally, rats are suggested as a preferred experimental model for studying S1P1 receptor-mediated responses in the human brain.
Collapse
Affiliation(s)
- Jonatan Martínez-Gardeazabal
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain; Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain
| | - Gorka Pereira-Castelo
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Marta Moreno-Rodríguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Alberto Llorente-Ovejero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Manuel Fernández
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain; Department of Neurology, Hospital Universitario de Cruces, 48903 Barakaldo, Spain
| | - Iván Fernández-Vega
- Department of Pathology, Hospital Universitario Central de Asturias, Avda. Roma, s/n, 33011 Oviedo, Spain; Health Research Institute of Principality of Asturias (ISPA), Av. del Hospital Universitario, s/n, 33011 Oviedo, Spain
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain; Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain.
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain; Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain
| |
Collapse
|
2
|
Farooqui AA, Farooqui T. Phospholipids, Sphingolipids, and Cholesterol-Derived Lipid Mediators and Their Role in Neurological Disorders. Int J Mol Sci 2024; 25:10672. [PMID: 39409002 PMCID: PMC11476704 DOI: 10.3390/ijms251910672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Neural membranes are composed of phospholipids, sphingolipids, cholesterol, and proteins. In response to cell stimulation or injury, the metabolism of lipids generates various lipid mediators, which perform many cellular functions. Thus, phospholipids release arachidonic acid or docosahexaenoic acid from the sn-2 position of the glycerol moiety by the action of phospholipases A2. Arachidonic acid is a precursor for prostaglandins, leukotrienes, thromboxane, and lipoxins. Among these mediators, prostaglandins, leukotrienes, and thromboxane produce neuroinflammation. In contrast, lipoxins produce anti-inflammatory and pro-resolving effects. Prostaglandins, leukotrienes, and thromboxane are also involved in cell proliferation, differentiation, blood clotting, and blood vessel permeability. In contrast, DHA-derived lipid mediators are called specialized pro-resolving lipid metabolites (SPMs). They include resolvins, protectins, and maresins. These mediators regulate immune function by producing anti-inflammatory, pro-resolving, and cell protective effects. Sphingolipid-derived metabolites are ceramide, ceramide1-phosphate, sphingosine, and sphingosine 1 phosphate. They regulate many cellular processes, including enzyme activities, cell migration and adhesion, inflammation, and immunity. Cholesterol is metabolized into hydroxycholesterols and 7-ketocholesterol, which not only disrupts membrane fluidity, but also promotes inflammation, oxidative stress, and apoptosis. These processes lead to cellular damage.
Collapse
Affiliation(s)
| | - Tahira Farooqui
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
3
|
Thonhofer M, Culum A, Dorn T, Fischer R, Prasch H, Stütz AE, Weber P, Wrodnigg TM. A concise synthetic approach for isoiminosugars. Carbohydr Res 2024; 544:109239. [PMID: 39142016 DOI: 10.1016/j.carres.2024.109239] [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: 06/27/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Isoiminosugars are highly biological active substances. Herein, we report a concise synthetic approach for this class of compounds. The key step relies on a stereospecific 1,2-hydride shift in O-2 tosylated glycopyranosides leading to C-2 branched glycofuranosides. This approach enables a 4-step synthesis of powerful β-galactosidase inhibitor 4-epi-isofagomine starting from a simple d-glucopyranoside.
Collapse
Affiliation(s)
- Martin Thonhofer
- Graz University of Technology, Institute of Chemistry and Technology of Biobased Systems, Stremayrgasse 9, A-8010, Graz, Austria.
| | - André Culum
- Graz University of Technology, Institute of Chemistry and Technology of Biobased Systems, Stremayrgasse 9, A-8010, Graz, Austria
| | - Tobias Dorn
- Graz University of Technology, Institute of Chemistry and Technology of Biobased Systems, Stremayrgasse 9, A-8010, Graz, Austria
| | - Roland Fischer
- Graz University of Technology, Institute of Inorganic Chemistry, Stremayrgasse 9, A-8010, Graz, Austria
| | - Herwig Prasch
- Graz University of Technology, Institute of Chemistry and Technology of Biobased Systems, Stremayrgasse 9, A-8010, Graz, Austria
| | - Arnold E Stütz
- Graz University of Technology, Institute of Chemistry and Technology of Biobased Systems, Stremayrgasse 9, A-8010, Graz, Austria
| | - Patrick Weber
- Graz University of Technology, Institute of Chemistry and Technology of Biobased Systems, Stremayrgasse 9, A-8010, Graz, Austria
| | - Tanja M Wrodnigg
- Graz University of Technology, Institute of Chemistry and Technology of Biobased Systems, Stremayrgasse 9, A-8010, Graz, Austria
| |
Collapse
|
4
|
Yan Y, Gao Y, Kumar G, Fang Q, Yan H, Zhang N, Zhang Y, Song L, Li J, Zheng Y, Zhang N, Zhang P, Ma C. Exosomal MicroRNAs modulate the cognitive function in fasudil treated APPswe/PSEN1dE9 transgenic (APP/PS1) mice model of Alzheimer's disease. Metab Brain Dis 2024; 39:1335-1351. [PMID: 39088109 PMCID: PMC11513711 DOI: 10.1007/s11011-024-01395-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline stemming from the accumulation of beta-amyloid (Aβ) plaques and the propagation of tau pathology through synapses. Exosomes, crucial mediators in neuronal development, maintenance, and intercellular communication, have gained attention in AD research. Yet, the molecular mechanisms involving exosomal miRNAs in AD remain elusive. In this study, we treated APPswe/PSEN1dE9 transgenic (APP/PS1) mice, a model for AD, with either vehicle (ADNS) or fasudil (ADF), while C57BL/6 (control) mice received vehicle (WT). Cognitive function was evaluated using the Y-maze test, and AD pathology was confirmed through immunostaining and western blot analysis of Aβ plaques and phosphorylated tau. Exosomal RNAs were extracted, sequenced, and analyzed from each mouse group. Our findings revealed that fasudil treatment improved cognitive function in AD mice, as evidenced by increased spontaneous alternation in the Y-maze test and reduced Aβ plaque load and phosphorylated tau protein expression in the hippocampus. Analysis of exosomal miRNAs identified three miRNAs (mmu-let-7i-5p, mmu-miR-19a-3p, mmu-miR-451a) common to both ADNS vs ADF and WT vs ADNS groups. Utilizing miRTarBase software, we predicted and analyzed target genes associated with these miRNAs. Gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of miRNA target genes indicated that mmu-miR-19a-3p and mmu-miR-451a are implicated in signal transduction, immune response, cellular communication, and nervous system pathways. Specifically, mmu-miR-19a-3p targeted genes involved in the sphingolipid signaling pathway, such as Pten and Tnf, while mmu-miR-451a targeted Nsmaf, Gnai3, and Akt3. Moreover, mmu-miR-451a targeted Myc in signaling pathways regulating the pluripotency of stem cells. In conclusion, fasudil treatment enhanced cognitive function by modulating exosomal MicroRNAs, particularly mmu-miR-451a and mmu-miR-19a-3p. These miRNAs hold promise as potential biomarkers and therapeutic targets for novel AD treatments.
Collapse
Affiliation(s)
- Yuqing Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
| | - Ye Gao
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong.
| | - Qingli Fang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Hailong Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Nianping Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Yuna Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jiehui Li
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Yucheng Zheng
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Nan Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Peijun Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Cungen Ma
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple, Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China.
| |
Collapse
|
5
|
Güleç A, Türkoğlu S, Kocabaş R. The relationship between sphingomyelin and ceramide levels and soft neurological signs in ADHD. J Neural Transm (Vienna) 2024:10.1007/s00702-024-02831-w. [PMID: 39249516 DOI: 10.1007/s00702-024-02831-w] [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: 04/09/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
Attention deficit hyperactivity disorder (ADHD), characterized by attention deficit, hyperactivity, and impulsivity, has recently been associated with lipid metabolism. In particular, the roles of sphingomyelin, ceramide, andgalactosylceramidase in the pathophysiology of ADHD are being investigated. This study aims to explore the relationship between sphingolipid metabolism markers and soft neurological signs (SNS) in children diagnosed with ADHD who are not undergoing medication treatment. A cross-sectional analysis was conducted on 41 children and adolescents aged 7-12 years diagnosed with ADHD and 39 neurotypically developing controls. Plasma levels of ceramide, sphingomyelin, and galactosylceramidase were measuredusing Enzyme-Linked Immunosorbent Assay (ELISA). SNS were assessed using the Physical and Neurological Examination for Soft Signs (PANESS). Statistical analyses included Student's t-tests, Mann-Whitney U tests, and Multivariate Analysis ofCovariance (MANCOVA), along with logistic regression analysis. Plasma levels of ceramide and sphingomyelin in children with ADHD showed significant differences compared to the neurotypically developing control group; however, there were no significant differences in galactosylceramidase levels between the two groups. Positive correlations were found between plasma levels of ceramide and sphingomyelin and the PANESS subscales F1 (Total Gait and Station) and F3 (Total Dysrhythmia). Additionally, logistic regression analysis indicated that high ceramide levels were positively associated with ADHD. This study underscores a significant association between alterations in sphingolipid metabolism (specifically increased levels of ceramide and sphingomyelin) and the presence of SNS in children with ADHD. These findings elucidate the potential role of sphingolipid metabolism in the pathophysiology of ADHD and provide suggestions for future therapeutic research targeting sphingolipid metabolism in the treatment of ADHD.
Collapse
Affiliation(s)
- Ahmet Güleç
- Department of Child and Adolescent Psychiatry, 209th Sk. No:26, 10100 Altıeylül/Balıkesir -Balıkesir Ataturk City Hospital, Gaziosmanpasa, Turkey.
| | - Serhat Türkoğlu
- Department of Child and Adolescent Psychiatry, Selcuk University Faculty of Medicine, Selçuk University Rectorate, Alaeddin Keykubat Campus, Academia District, New Istanbul Street No: 369, Selçuklu-Konya, Postal Code: 42130, Turkey
| | - Ramazan Kocabaş
- Department of Biochemistry, Selcuk University Faculty of Medicine, Selcuk University, Selçuk University Rectorate, Alaeddin Keykubat Campus, Academia District, New Istanbul Street No: 369, Selçuklu-Konya, Postal Code: 42130, Turkey
| |
Collapse
|
6
|
Roytrakul S, Jaresitthikunchai J, Phaonakrop N, Charoenlappanit S, Thaisakun S, Kumsri N, Arpornsuwan T. Secretomic changes of amyloid beta peptides on Alzheimer's disease related proteins in differentiated human SH-SY5Y neuroblastoma cells. PeerJ 2024; 12:e17732. [PMID: 39035166 PMCID: PMC11260076 DOI: 10.7717/peerj.17732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that causes physical damage to neuronal connections, leading to brain atrophy. This disruption of synaptic connections results in mild to severe cognitive impairments. Unfortunately, no effective treatment is currently known to prevent or reverse the symptoms of AD. The aim of this study was to investigate the effects of three synthetic peptides, i.e., KLVFF, RGKLVFFGR and RIIGL, on an AD in vitro model represented by differentiated SH-SY5Y neuroblastoma cells exposed to retinoic acid (RA) and brain-derived neurotrophic factor (BDNF). The results demonstrated that RIIGL peptide had the least significant cytotoxic activity to normal SH-SY5Y while exerting high cytotoxicity against the differentiated cells. The mechanism of RIIGL peptide in the differentiated SH-SY5Y was investigated based on changes in secretory proteins compared to another two peptides. A total of 380 proteins were identified, and five of them were significantly detected after treatment with RIIGL peptide. These secretory proteins were found to be related to microtubule-associated protein tau (MAPT) and amyloid-beta precursor protein (APP). RIIGL peptide acts on differentiated SH-SY5Y by regulating amyloid-beta formation, neuron apoptotic process, ceramide catabolic process, and oxidative phosphorylation and thus has the potentials to treat AD.
Collapse
Affiliation(s)
- Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Janthima Jaresitthikunchai
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Narumon Phaonakrop
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sawanya Charoenlappanit
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Siriwan Thaisakun
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Nitithorn Kumsri
- Undergraduate Student of Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumtani, Thailand
| | - Teerakul Arpornsuwan
- Medical Technology Research and Service Unit, Health Care Service Center, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
7
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of pathology in a mouse model of Alzheimer's disease. Nat Commun 2024; 15:5217. [PMID: 38890307 PMCID: PMC11189507 DOI: 10.1038/s41467-024-49589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jessica H Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
8
|
Ellis RJ, Pal S, Achim CL, Sundermann E, Moore DJ, Soontornniyomkij V, Feldman H. Alzheimer-Type Cerebral Amyloidosis in the Context of HIV Infection: Implications for a Proposed New Treatment Approach. J Neuroimmune Pharmacol 2024; 19:27. [PMID: 38829507 PMCID: PMC11147830 DOI: 10.1007/s11481-024-10126-w] [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: 06/08/2023] [Accepted: 05/13/2024] [Indexed: 06/05/2024]
Abstract
Reverse transcriptase inhibitors (RTIs) are currently broadly prescribed for the treatment of HIV infection but are also thought to prevent Alzheimer's disease (AD) progression by protecting against amyloidosis. Our study evaluates the hypothesis that reverse transcriptase inhibitors protect against Alzheimer-type brain amyloidogenesis in the context of HIV infection. We compiled a case series of participants from a prospective study of the neurological consequences of HIV infection at the HIV Neurobehavioral Research Program (HNRP) who had serial neuropsychological and neurological assessments and were on RTIs. Two participants had gross and microscopic examination and immunohistochemistry of the brain at autopsy; one was assessed clinically for Alzheimer's disease by cerebrospinal fluid (CSF) analysis of phosphorylated-Tau, Total-Tau and Aβ42. Additionally, a larger cohort of 250 autopsied individuals was evaluated for presence of amyloid plaques, Tau, and related pathologies. Three older, virally suppressed individuals with HIV who had long-term treatment with RTIs were included in analyses. Two cases demonstrated substantial cerebral amyloid deposition at autopsy. The third case met clinical criteria for AD based on a typical clinical course and CSF biomarker profile. In the larger cohort of autopsied individuals, the prevalence of cerebral amyloidosis among people with HIV (PWH) was greater for those on RTIs. Our study showed that long-term RTI therapy did not protect against Alzheimer-type brain amyloidogenesis in the context of HIV infection in these patients. Given the known toxicities of RTIs, it is premature to recommend them to individuals at risk or with Alzheimer's disease who do not have HIV infection.
Collapse
Affiliation(s)
- Ronald J Ellis
- Department of Neuroscience, University of California, San Diego, CA, USA.
- Department of Psychiatry, University of California, San Diego, CA, USA.
| | - Shibangi Pal
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Cristian L Achim
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Erin Sundermann
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - David J Moore
- Department of Psychiatry, University of California, San Diego, CA, USA
| | | | - Howard Feldman
- Department of Neuroscience, University of California, San Diego, CA, USA
| |
Collapse
|
9
|
Su H, Masters CL, Bush AI, Barnham KJ, Reid GE, Vella LJ. Exploring the significance of lipids in Alzheimer's disease and the potential of extracellular vesicles. Proteomics 2024; 24:e2300063. [PMID: 37654087 DOI: 10.1002/pmic.202300063] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
Lipids play a significant role in maintaining central nervous system (CNS) structure and function, and the dysregulation of lipid metabolism is known to occur in many neurological disorders, including Alzheimer's disease. Here we review what is currently known about lipid dyshomeostasis in Alzheimer's disease. We propose that small extracellular vesicle (sEV) lipids may provide insight into the pathophysiology and progression of Alzheimer's disease. This stems from the recognition that sEV likely contributes to disease pathogenesis, but also an understanding that sEV can serve as a source of potential biomarkers. While the protein and RNA content of sEV in the CNS diseases have been studied extensively, our understanding of the lipidome of sEV in the CNS is still in its infancy.
Collapse
Affiliation(s)
- Huaqi Su
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin J Barnham
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
| | - Gavin E Reid
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura J Vella
- The Florey, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
11
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
12
|
Jo T, Kim J, Bice P, Huynh K, Wang T, Arnold M, Meikle PJ, Giles C, Kaddurah-Daouk R, Saykin AJ, Nho K. Circular-SWAT for deep learning based diagnostic classification of Alzheimer's disease: application to metabolome data. EBioMedicine 2023; 97:104820. [PMID: 37806288 PMCID: PMC10579282 DOI: 10.1016/j.ebiom.2023.104820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND Deep learning has shown potential in various scientific domains but faces challenges when applied to complex, high-dimensional multi-omics data. Alzheimer's Disease (AD) is a neurodegenerative disorder that lacks targeted therapeutic options. This study introduces the Circular-Sliding Window Association Test (c-SWAT) to improve the classification accuracy in predicting AD using serum-based metabolomics data, specifically lipidomics. METHODS The c-SWAT methodology builds upon the existing Sliding Window Association Test (SWAT) and utilizes a three-step approach: feature correlation analysis, feature selection, and classification. Data from 997 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) served as the basis for model training and validation. Feature correlations were analyzed using Weighted Gene Co-expression Network Analysis (WGCNA), and Convolutional Neural Networks (CNN) were employed for feature selection. Random Forest was used for the final classification. FINDINGS The application of c-SWAT resulted in a classification accuracy of up to 80.8% and an AUC of 0.808 for distinguishing AD from cognitively normal older adults. This marks a 9.4% improvement in accuracy and a 0.169 increase in AUC compared to methods without c-SWAT. These results were statistically significant, with a p-value of 1.04 × 10ˆ-4. The approach also identified key lipids associated with AD, such as Cer(d16:1/22:0) and PI(37:6). INTERPRETATION Our results indicate that c-SWAT is effective in improving classification accuracy and in identifying potential lipid biomarkers for AD. These identified lipids offer new avenues for understanding AD and warrant further investigation. FUNDING The specific funding of this article is provided in the acknowledgements section.
Collapse
Affiliation(s)
- Taeho Jo
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Junpyo Kim
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Medical Research Institute, Sungkyunkwan University, School of Medicine, Seoul, South Korea
| | - Paula Bice
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Tingting Wang
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, 27710, USA; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, 3010, Victoria, Australia; Monash University, Melbourne, VIC 3800, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, 27710, USA; Duke Institute of Brain Sciences, Duke University, Durham, NC, 27710, USA; Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
13
|
Pihlström S, Richardt S, Määttä K, Pekkinen M, Olkkonen VM, Mäkitie O, Mäkitie RE. SGMS2 in primary osteoporosis with facial nerve palsy. Front Endocrinol (Lausanne) 2023; 14:1224318. [PMID: 37886644 PMCID: PMC10598846 DOI: 10.3389/fendo.2023.1224318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
Pathogenic heterozygous variants in SGMS2 cause a rare monogenic form of osteoporosis known as calvarial doughnut lesions with bone fragility (CDL). The clinical presentations of SGMS2-related bone pathology range from childhood-onset osteoporosis with low bone mineral density and sclerotic doughnut-shaped lesions in the skull to a severe spondylometaphyseal dysplasia with neonatal fractures, long-bone deformities, and short stature. In addition, neurological manifestations occur in some patients. SGMS2 encodes sphingomyelin synthase 2 (SMS2), an enzyme involved in the production of sphingomyelin (SM). This review describes the biochemical structure of SM, SM metabolism, and their molecular actions in skeletal and neural tissue. We postulate how disrupted SM gradient can influence bone formation and how animal models may facilitate a better understanding of SGMS2-related osteoporosis.
Collapse
Affiliation(s)
- Sandra Pihlström
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sampo Richardt
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi Määttä
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Minna Pekkinen
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children´s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Vesa M. Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Outi Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children´s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Riikka E. Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Otorhinolaryngology – Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
14
|
Obis E, Sol J, Andres-Benito P, Martín-Gari M, Mota-Martorell N, Galo-Licona JD, Piñol-Ripoll G, Portero-Otin M, Ferrer I, Jové M, Pamplona R. Lipidomic Alterations in the Cerebral Cortex and White Matter in Sporadic Alzheimer's Disease. Aging Dis 2023; 14:1887-1916. [PMID: 37196109 PMCID: PMC10529741 DOI: 10.14336/ad.2023.0217] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/17/2023] [Indexed: 05/19/2023] Open
Abstract
Non-targeted LC-MS/MS-based lipidomic analysis was conducted in post-mortem human grey matter frontal cortex area 8 (GM) and white matter of the frontal lobe centrum semi-ovale (WM) to identify lipidome fingerprints in middle-aged individuals with no neurofibrillary tangles and senile plaques, and cases at progressive stages of sporadic Alzheimer's disease (sAD). Complementary data were obtained using RT-qPCR and immunohistochemistry. The results showed that WM presents an adaptive lipid phenotype resistant to lipid peroxidation, characterized by a lower fatty acid unsaturation, peroxidizability index, and higher ether lipid content than the GM. Changes in the lipidomic profile are more marked in the WM than in GM in AD with disease progression. Four functional categories are associated with the different lipid classes affected in sAD: membrane structural composition, bioenergetics, antioxidant protection, and bioactive lipids, with deleterious consequences affecting both neurons and glial cells favoring disease progression.
Collapse
Affiliation(s)
- Elia Obis
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Joaquim Sol
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
- Catalan Institute of Health (ICS), Lleida, Spain, Research Support Unit (USR), Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol), Lleida, Spain.
| | - Pol Andres-Benito
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.
- Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), E-08907 Hospitalet de Llobregat, Barcelona, Spain.
| | - Meritxell Martín-Gari
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Natàlia Mota-Martorell
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - José Daniel Galo-Licona
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Clinical Neuroscience Research, Santa Maria University Hospital, IRBLleida, Lleida, Spain.
| | - Manuel Portero-Otin
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Isidro Ferrer
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.
- Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), E-08907 Hospitalet de Llobregat, Barcelona, Spain.
- Department of Pathology and Experimental Therapeutics, University of Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain.
| | - Mariona Jové
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida University (UdL), Lleida Biomedical Research Institute (IRBLleida), Lleida, Spain.
| |
Collapse
|
15
|
Ellis RJ, Pal S, Achim CL, Sundermann E, Moore DJ, Soontornniyomkij V, Feldman H. Alzheimer-type cerebral amyloidosis in the context of HIV infection: implications for a proposed new treatment approach. RESEARCH SQUARE 2023:rs.3.rs-3040756. [PMID: 37398361 PMCID: PMC10312930 DOI: 10.21203/rs.3.rs-3040756/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Reverse transcriptase inhibitors (RTIs) are currently broadly prescribed for the treatment of HIV infection but are also thought to prevent Alzheimer's Disease (AD) progression by protecting against amyloidosis. Our study evaluates the hypothesis that reverse transcriptase inhibitors protect against Alzheimer-type brain amyloidogenesis in the context of HIV infection. We compiled a case series of participants from a prospective study of the neurological consequences of HIV infection at the HIV Neurobehavioral Research Program (HNRP) who had serial neuropsychological and neurological assessments and were on RTIs. Two participants had gross and microscopic examination and immunohistochemistry of the brain at autopsy; one was assessed clinically for Alzheimer's Disease by cerebrospinal fluid (CSF) analysis of phosphorylated-Tau, Total-Tau and Aβ42. Additionally, a larger cohort of autopsied individuals was evaluated for presence of amyloid plaques, Tau, and related pathologies. Three older, virally suppressed individuals with HIV who had long-term treatment with RTIs were included in analyses. Two cases demonstrated substantial cerebral amyloid deposition at autopsy. The third case met clinical criteria for AD based on a typical clinical course and CSF biomarker profile. In the larger cohort of autopsied individuals, the prevalence of cerebral amyloidosis among people with HIV (PWH) was greater for those on RTIs. Our study showed that long-term RTI therapy did not protect against Alzheimer-type brain amyloidogenesis in the context of HIV infection in these patients. Given the known toxicities of RTIs, it is premature to recommend them to individuals at risk or with Alzheimer's disease who do not have HIV infection.
Collapse
|
16
|
Min EK, Lee H, Sung EJ, Seo SW, Song M, Wang S, Kim SS, Bae MA, Kim TY, Lee S, Kim KT. Integrative multi-omics reveals analogous developmental neurotoxicity mechanisms between perfluorobutanesulfonic acid and perfluorooctanesulfonic acid in zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131714. [PMID: 37263023 DOI: 10.1016/j.jhazmat.2023.131714] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
The molecular mechanism of perfluorobutanesulfonic acid (PFBS), an alternative to legacy perfluorooctanesulfonic acid (PFOS), is not fully understood yet. Therefore, we conducted a developmental toxicity evaluation on zebrafish embryos exposed to PFBS and PFOS and assessed neurobehavioral changes at concentrations below each point of departure (POD) determined by embryonic mortality. Using transcriptomics, proteomics, and metabolomics, biomolecular perturbations in response to PFBS were profiled and then integrated for comparison with those for PFOS. Although PFBS (7525.47 μM POD) was approximately 700 times less toxic than PFOS (11.42 μM POD), altered neurobehavior patterns and affected kinds of endogenous neurochemicals were similar between PFBS and PFOS at the corresponding POD-based concentrations. Multi-omics analysis revealed that the PFBS neurotoxicity mechanism was associated with oxidative stress, lipid metabolism, and glycolysis/glucogenesis. The commonalities in developmental neurotoxicity-related mechanisms between PFBS and PFOS interconnected by knowledge-based integration of multi-omics included the calcium signaling pathway, lipid homeostasis, and primary bile acid biosynthesis. Despite being less toxic than PFOS, PFBS exhibited similar dysregulated molecular mechanisms, suggesting that chain length differences do not affect the intrinsic toxicity mechanism. Overall, carefully managing potential toxicity of PFBS can secure its status as an alternative to PFOS.
Collapse
Affiliation(s)
- Eun Ki Min
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Hyojin Lee
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Eun Ji Sung
- College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Seong Woo Seo
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Myungha Song
- Environmental Health Research Department, National Institute of Environmental Research, Incheon 22689, Republic of Korea
| | - Seungjun Wang
- Environmental Health Research Department, National Institute of Environmental Research, Incheon 22689, Republic of Korea
| | - Seong Soon Kim
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Tae-Young Kim
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - Sangkyu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Ki-Tae Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea.
| |
Collapse
|
17
|
den Hoedt S, Dorst-Lagerwerf KY, de Vries HE, Rozemuller AJ, Scheltens P, Walter J, Sijbrands EJ, Martinez-Martinez P, Verhoeven AJ, Teunissen CE, Mulder MT. Sphingolipids in Cerebrospinal Fluid and Plasma Lipoproteins of APOE4 Homozygotes and Non-APOE4 Carriers with Mild Cognitive Impairment versus Subjective Cognitive Decline. J Alzheimers Dis Rep 2023; 7:339-354. [DOI: 10.3233/adr220072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Background: Alzheimer’s disease (AD) patients display alterations in cerebrospinal fluid (CSF) and plasma sphingolipids. The APOE4 genotype increases the risk of developing AD. Objective: To test the hypothesis that the APOE4 genotype affects common sphingolipids in CSF and in plasma of patients with early stages of AD. Methods: Patients homozygous for APOE4 and non-APOE4 carriers with mild cognitive impairment (MCI; n = 20 versus 20) were compared to patients with subjective cognitive decline (SCD; n = 18 versus 20). Sphingolipids in CSF and plasma lipoproteins were determined by liquid-chromatography-tandem mass spectrometry. Aβ42 levels in CSF were determined by immunoassay. Results: APOE4 homozygotes displayed lower levels of sphingomyelin (SM; p = 0.042), SM(d18:1/18:0) (p = 0.026), and Aβ 42 (p < 0.001) in CSF than non-APOE4 carriers. CSF-Aβ 42 correlated with Cer(d18:1/18:0), SM(d18:1/18:0), and SM(d18:1/18:1) levels in APOE4 homozygotes (r > 0.49; p < 0.032) and with Cer(d18:1/24:1) in non-APOE4 carriers (r = 0.50; p = 0.025). CSF-Aβ 42 correlated positively with Cer(d18:1/24:0) in MCI (p = 0.028), but negatively in SCD patients (p = 0.019). Levels of Cer(d18:1/22:0) and long-chain SMs were inversely correlated with Mini-Mental State Examination score among MCI patients, independent of APOE4 genotype (r< –0.47; p < 0.039). Nevertheless, age and sex are stronger determinants of individual sphingolipid levels in CSF than either the APOE genotype or the cognitive state. In HDL, ratios of Cer(d18:1/18:0) and Cer(d18:1/22:0) to cholesterol were higher in APOE4 homozygotes than in non-APOE4 carriers (p = 0.048 and 0.047, respectively). Conclusion: The APOE4 genotype affects sphingolipid profiles of CSF and plasma lipoproteins already at early stages of AD. ApoE4 may contribute to the early development of AD through modulation of sphingolipid metabolism.
Collapse
Affiliation(s)
- Sandra den Hoedt
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, The Netherlands
| | - Annemieke J.M. Rozemuller
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Clinical Chemistry, The Alzheimer Center Amsterdam, and Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center, VrijeUniversiteit Amsterdam, The Netherlands
| | - Jochen Walter
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Eric J.G. Sijbrands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Pilar Martinez-Martinez
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Adrie J.M. Verhoeven
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Charlotte E. Teunissen
- Department of Clinical Chemistry, The Alzheimer Center Amsterdam, and Neurochemistry Laboratory, Amsterdam Neuroscience, Amsterdam University Medical Center, VrijeUniversiteit Amsterdam, The Netherlands
| | - Monique T. Mulder
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Burtscher J, Pepe G, Maharjan N, Riguet N, Di Pardo A, Maglione V, Millet GP. Sphingolipids and impaired hypoxic stress responses in Huntington disease. Prog Lipid Res 2023; 90:101224. [PMID: 36898481 DOI: 10.1016/j.plipres.2023.101224] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/20/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
Huntington disease (HD) is a debilitating, currently incurable disease. Protein aggregation and metabolic deficits are pathological hallmarks but their link to neurodegeneration and symptoms remains debated. Here, we summarize alterations in the levels of different sphingolipids in an attempt to characterize sphingolipid patterns specific to HD, an additional molecular hallmark of the disease. Based on the crucial role of sphingolipids in maintaining cellular homeostasis, the dynamic regulation of sphingolipids upon insults and their involvement in cellular stress responses, we hypothesize that maladaptations or blunted adaptations, especially following cellular stress due to reduced oxygen supply (hypoxia) contribute to the development of pathology in HD. We review how sphingolipids shape cellular energy metabolism and control proteostasis and suggest how these functions may fail in HD and in combination with additional insults. Finally, we evaluate the potential of improving cellular resilience in HD by conditioning approaches (improving the efficiency of cellular stress responses) and the role of sphingolipids therein. Sphingolipid metabolism is crucial for cellular homeostasis and for adaptations following cellular stress, including hypoxia. Inadequate cellular management of hypoxic stress likely contributes to HD progression, and sphingolipids are potential mediators. Targeting sphingolipids and the hypoxic stress response are novel treatment strategies for HD.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| | - Giuseppe Pepe
- IRCCS Neuromed, Via Dell'Elettronica, 86077 Pozzilli, Italy
| | - Niran Maharjan
- Department of Neurology, Center for Experimental Neurology, Inselspital University Hospital, 3010 Bern, Switzerland; Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland
| | | | - Alba Di Pardo
- IRCCS Neuromed, Via Dell'Elettronica, 86077 Pozzilli, Italy
| | | | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
19
|
Tohumeken S, Deme P, Yoo SW, Gupta S, Rais R, Slusher BS, Haughey NJ. Neuronal deletion of nSMase2 reduces the production of Aβ and directly protects neurons. Neurobiol Dis 2023; 177:105987. [PMID: 36603748 DOI: 10.1016/j.nbd.2023.105987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/03/2023] Open
Abstract
Extracellular vesicles (EVs) have been proposed to regulate the deposition of Aβ. Multiple publications have shown that APP, amyloid processing enzymes and Aβ peptides are associated with EVs. However, very little Aβ is associated with EVs compared with the total amount Aβ present in human plasma, CSF, or supernatants from cultured neurons. The involvement of EVs has largely been inferred by pharmacological inhibition or whole body deletion of the sphingomyelin hydrolase neutral sphingomyelinase-2 (nSMase2) that is a key regulator for the biogenesis of at-least one population of EVs. Here we used a Cre-Lox system to selectively delete nSMase2 from pyramidal neurons in APP/PS1 mice (APP/PS1-SMPD3-Nex1) and found a ∼ 70% reduction in Aβ deposition at 6 months of age and ∼ 35% reduction at 12 months of age in both cortex and hippocampus. Brain ceramides were increased in APP/PS1 compared with Wt mice, but were similar to Wt in APP/PS1-SMPD3-Nex1 mice suggesting that elevated brain ceramides in this model involves neuronally expressed nSMase2. Reduced levels of PSD95 and deficits of long-term potentiation in APP/PS1 mice were normalized in APP/PS1-SMPD3-Nex1 mice. In contrast, elevated levels of IL-1β, IL-8 and TNFα in APP/PS1 mice were not normalized in APP/PS1-SMPD3-Nex1 mice compared with APP/PS1 mice. Mechanistic studies showed that the size of liquid ordered membrane microdomains was increased in APP/PS1 mice, as were the amounts of APP and BACE1 localized to these microdomains. Pharmacological inhibition of nSMase2 activity with PDDC reduced the size of the liquid ordered membrane microdomains, reduced the localization of APP with BACE1 and reduced the production of Aβ1-40 and Aβ1-42. Although inhibition of nSMase2 reduced the release and increased the size of EVs, very little Aβ was associated with EVs in all conditions tested. We also found that nSMase2 directly protected neurons from the toxic effects of oligomerized Aβ and preserved neural network connectivity despite considerable Aβ deposition. These data demonstrate that nSMase2 plays a role in the production of Aβ by stabilizing the interaction of APP with BACE1 in liquid ordered membrane microdomains, and directly protects neurons from the toxic effects of Aβ. The effects of inhibiting nSMase2 on EV biogenesis may be independent from effects on Aβ production and neuronal protection.
Collapse
Affiliation(s)
- Sehmus Tohumeken
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Pragney Deme
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Seung Wan Yoo
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Sujasha Gupta
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Rana Rais
- The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America
| | - Barbara S Slusher
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America; The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America; The Johns Hopkins University School of Medicine, Departments of Pharmacology and Molecular Sciences, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Oncology, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Neuroscience, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Medicine, Baltimore, MD, United States of America
| | - Norman J Haughey
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America.
| |
Collapse
|
20
|
Mani S, Dubey R, Lai IC, Babu MA, Tyagi S, Swargiary G, Mody D, Singh M, Agarwal S, Iqbal D, Kumar S, Hamed M, Sachdeva P, Almutary AG, Albadrani HM, Ojha S, Singh SK, Jha NK. Oxidative Stress and Natural Antioxidants: Back and Forth in the Neurological Mechanisms of Alzheimer's Disease. J Alzheimers Dis 2023; 96:877-912. [PMID: 37927255 DOI: 10.3233/jad-220700] [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] [Indexed: 11/07/2023]
Abstract
Alzheimer's disease (AD) is characterized by the progressive degeneration of neuronal cells. With the increase in aged population, there is a prevalence of irreversible neurodegenerative changes, causing a significant mental, social, and economic burden globally. The factors contributing to AD are multidimensional, highly complex, and not completely understood. However, it is widely known that aging, neuroinflammation, and excessive production of reactive oxygen species (ROS), along with other free radicals, substantially contribute to oxidative stress and cell death, which are inextricably linked. While oxidative stress is undeniably important in AD, limiting free radicals and ROS levels is an intriguing and potential strategy for deferring the process of neurodegeneration and alleviating associated symptoms. Therapeutic compounds from natural sources have recently become increasingly accepted and have been effectively studied for AD treatment. These phytocompounds are widely available and a multitude of holistic therapeutic efficiencies for treating AD owing to their antioxidant, anti-inflammatory, and biological activities. Some of these compounds also function by stimulating cholinergic neurotransmission, facilitating the suppression of beta-site amyloid precursor protein-cleaving enzyme 1, α-synuclein, and monoamine oxidase proteins, and deterring the occurrence of AD. Additionally, various phenolic, flavonoid, and terpenoid phytocompounds have been extensively described as potential palliative agents for AD progression. Preclinical studies have shown their involvement in modulating the cellular redox balance and minimizing ROS formation, displaying them as antioxidant agents with neuroprotective abilities. This review emphasizes the mechanistic role of natural products in the treatment of AD and discusses the various pathological hypotheses proposed for AD.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Rajni Dubey
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - I-Chun Lai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Sakshi Tyagi
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Deepansh Mody
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Manisha Singh
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Shriya Agarwal
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah, Saudi Arabia
| | - Sanjay Kumar
- Department of Life Sciences, School of Basic Sciences and Research (SBSR), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Munerah Hamed
- Department of Pathology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Hind Muteb Albadrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Kingdom of Saudi Arabia
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | | | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, Uttarakhand, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
| |
Collapse
|
21
|
Corrigan RR, Labrador L, Grizzanti J, Mey M, Piontkivska H, Casadesús G. Neuroprotective Mechanisms of Amylin Receptor Activation, Not Antagonism, in the APP/PS1 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2023; 91:1495-1514. [PMID: 36641678 DOI: 10.3233/jad-221057] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Amylin, a pancreatic amyloid peptide involved in energy homeostasis, is increasingly studied in the context of Alzheimer's disease (AD) etiology. To date, conflicting pathogenic and neuroprotective roles for this peptide and its analogs for AD pathogenesis have been described. OBJECTIVE Whether the benefits of amylin are associated with peripheral improvement of metabolic tone/function or directly through the activation of central amylin receptors is also unknown and downstream signaling mechanisms of amylin receptors are major objectives of this study. METHODS To address these questions more directly we delivered the amylin analog pramlintide systemically (IP), at previously identified therapeutic doses, while centrally (ICV) inhibiting the receptor using an amylin receptor antagonist (AC187), at doses known to impact CNS function. RESULTS Here we show that pramlintide improved cognitive function independently of CNS receptor activation and provide transcriptomic data that highlights potential mechanisms. Furthermore, we show than inhibition of the amylin receptor increased amyloid-beta pathology in female APP/PS1 mice, an effect than was mitigated by peripheral delivery of pramlintide. Through transcriptomic analysis of pramlintide therapy in AD-modeled mice we found sexual dimorphic modulation of neuroprotective mechanisms: oxidative stress protection in females and membrane stability and reduced neuronal excitability markers in males. CONCLUSION These data suggest an uncoupling of functional and pathology-related events and highlighting a more complex receptor system and pharmacological relationship that must be carefully studied to clarify the role of amylin in CNS function and AD.
Collapse
Affiliation(s)
| | - Luis Labrador
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - John Grizzanti
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Megan Mey
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Helen Piontkivska
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Gemma Casadesús
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
22
|
Mandik F, Kanana Y, Rody J, Misera S, Wilken B, Laabs von Holt BH, Klein C, Vos M. A new model for fatty acid hydroxylase-associated neurodegeneration reveals mitochondrial and autophagy abnormalities. Front Cell Dev Biol 2022; 10:1000553. [PMID: 36589738 PMCID: PMC9794614 DOI: 10.3389/fcell.2022.1000553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
Fatty acid hydroxylase-associated neurodegeneration (FAHN) is a rare disease that exhibits brain modifications and motor dysfunctions in early childhood. The condition is caused by a homozygous or compound heterozygous mutation in fatty acid 2 hydroxylase (FA2H), whose encoded protein synthesizes 2-hydroxysphingolipids and 2-hydroxyglycosphingolipids and is therefore involved in sphingolipid metabolism. A few FAHN model organisms have already been established and give the first insight into symptomatic effects. However, they fail to establish the underlying cellular mechanism of FAHN so far. Drosophila is an excellent model for many neurodegenerative disorders; hence, here, we have characterized and validated the first FAHN Drosophila model. The investigation of loss of dfa2h lines revealed behavioral abnormalities, including motor impairment and flying disability, in addition to a shortened lifespan. Furthermore, alterations in mitochondrial dynamics, and autophagy were identified. Analyses of patient-derived fibroblasts, and rescue experiments with human FA2H, indicated that these defects are evolutionarily conserved. We thus present a FAHN Drosophila model organism that provides new insights into the cellular mechanism of FAHN.
Collapse
Affiliation(s)
- Frida Mandik
- Institute of Neurogenetics, University of Luebeck, UKSH, Luebeck, Germany
| | - Yuliia Kanana
- Institute of Neurogenetics, University of Luebeck, UKSH, Luebeck, Germany
| | - Jost Rody
- Institute of Neurogenetics, University of Luebeck, UKSH, Luebeck, Germany
| | - Sophie Misera
- Institute of Neurogenetics, University of Luebeck, UKSH, Luebeck, Germany
| | - Bernd Wilken
- Department of Neuropediatrics, Klinikum Kassel, Kassel, Germany
| | | | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, UKSH, Luebeck, Germany
| | - Melissa Vos
- Institute of Neurogenetics, University of Luebeck, UKSH, Luebeck, Germany,*Correspondence: Melissa Vos,
| |
Collapse
|
23
|
Li H, Tan Y, Cheng X, Zhang Z, Huang J, Hui S, Zhu L, Liu Y, Zhao D, Liu Z, Peng W. Untargeted metabolomics analysis of the hippocampus and cerebral cortex identified the neuroprotective mechanisms of Bushen Tiansui formula in an aβ25-35-induced rat model of Alzheimer’s disease. Front Pharmacol 2022; 13:990307. [PMID: 36339577 PMCID: PMC9630565 DOI: 10.3389/fphar.2022.990307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Bushen Tiansui Formula (BSTSF) is a traditional formulation of Chinese medicine that has been used to treat Alzheimer’s disease (AD) for decades; however, the underlying mechanisms by which this formula achieves such therapeutic effects have yet to be elucidated. Prupose: To investigate the neuroprotective mechanisms of BSTSF against AD by analyzing metabolite profiles in the hippocampus and cortex of AD rats. Methods: The rat models of AD were established by the injection of Aβ25–35. The Morris water maze (MWM) test was performed to evaluate the effect of BSTSF treatment on cognitive dysfunction. Hematoxylin and eosin (HE) staining was used to assess the effect of BSTSF on typical AD pathologies. Underlying mechanisms were investigated using LC-MS/MS-based untargeted metabolomics analysis of the cerebral cortex and hippocampus. Results: BSTSF significantly improved memory deficits and the typical histopathological changes of AD rats. Untargeted metabolomics analysis showed that 145 and 184 endogenous metabolites in the cerebral cortex and hippocampus, respectively, were significantly different in the BSTSF group when compared with the AD group. The differential metabolites in the cerebral cortex were primarily involved in cysteine and methionine metabolism, while those in the hippocampus were mainly involved in d-Glutamine and d-glutamate metabolism. Conclusion: In the present study, we confirmed the neuroprotective effects of BSTSF treatment against AD using a rat model. Our findings indicate that the BSTSF-mediated protective effects were associated with amelioration of metabolic disorders in the hippocampus and cerebral cortex.
Collapse
Affiliation(s)
- Hongli Li
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Mental Disorder, Changsha, China
| | - Yejun Tan
- School of mathematics, University of Minnesota twin Cities, St. Paul, MS, United States
| | - Xin Cheng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Mental Disorder, Changsha, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Mental Disorder, Changsha, China
| | | | - Shan Hui
- Department of Geratology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Yuqing Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Di Zhao
- Hunan Academy of Chinese Medicine, Changsha, China
| | - Zhao Liu
- Hunan Academy of Chinese Medicine, Changsha, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Mental Disorder, Changsha, China
- *Correspondence: Weijun Peng,
| |
Collapse
|
24
|
Martín MG, Dotti CG. Plasma membrane and brain dysfunction of the old: Do we age from our membranes? Front Cell Dev Biol 2022; 10:1031007. [PMID: 36274849 PMCID: PMC9582647 DOI: 10.3389/fcell.2022.1031007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022] Open
Abstract
One of the characteristics of aging is a gradual hypo-responsiveness of cells to extrinsic stimuli, mainly evident in the pathways that are under hormone control, both in the brain and in peripheral tissues. Age-related resistance, i.e., reduced response of receptors to their ligands, has been shown to Insulin and also to leptin, thyroid hormones and glucocorticoids. In addition, lower activity has been reported in aging for ß-adrenergic receptors, adenosine A2B receptor, and several other G-protein-coupled receptors. One of the mechanisms proposed to explain the loss of sensitivity to hormones and neurotransmitters with age is the loss of receptors, which has been observed in several tissues. Another mechanism that is finding more and more experimental support is related to the changes that occur with age in the lipid composition of the neuronal plasma membrane, which are responsible for changes in the receptors’ coupling efficiency to ligands, signal attenuation and pathway desensitization. In fact, recent works have shown that altered membrane composition—as occurs during neuronal aging—underlies reduced response to glutamate, to the neurotrophin BDNF, and to insulin, all these leading to cognition decay and epigenetic alterations in the old. In this review we present evidence that altered functions of membrane receptors due to altered plasma membrane properties may be a triggering factor in physiological decline, decreased brain function, and increased vulnerability to neuropathology in aging.
Collapse
Affiliation(s)
- Mauricio G. Martín
- Cellular and Molecular Neurobiology Department, Instituto Ferreyra (INIMEC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
- *Correspondence: Mauricio G. Martín, ; Carlos G. Dotti,
| | - Carlos G. Dotti
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- *Correspondence: Mauricio G. Martín, ; Carlos G. Dotti,
| |
Collapse
|
25
|
Banaras S, Paracha RZ, Nisar M, Arif A, Ahmad J, Tariq Saeed M, Mustansar Z, Shuja MN, Paracha RN. System level modeling and analysis of TNF- α mediated sphingolipid signaling pathway in neurological disorders for the prediction of therapeutic targets. Front Physiol 2022; 13:872421. [PMID: 36060699 PMCID: PMC9437628 DOI: 10.3389/fphys.2022.872421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/15/2022] [Indexed: 01/09/2023] Open
Abstract
Sphingomyelin (SM) belongs to a class of lipids termed sphingolipids. The disruption in the sphingomyelin signaling pathway is associated with various neurodegenerative disorders. TNF-α, a potent pro-inflammatory cytokine generated in response to various neurological disorders like Alzheimer's disease (AD), Parkinson's disease (PD), and Multiple Sclerosis (MS), is an eminent regulator of the sphingomyelin metabolic pathway. The immune-triggered regulation of the sphingomyelin metabolic pathway via TNF-α constitutes the sphingomyelin signaling pathway. In this pathway, sphingomyelin and its downstream sphingolipids activate various signaling cascades like PI3K/AKT and MAPK/ERK pathways, thus, controlling diverse processes coupled with neuronal viability, survival, and death. The holistic analysis of the immune-triggered sphingomyelin signaling pathway is imperative to make necessary predictions about its pivotal components and for the formulation of disease-related therapeutics. The current work offers a comprehensive in silico systems analysis of TNF-α mediated sphingomyelin and downstream signaling cascades via a model-based quantitative approach. We incorporated the intensity values of genes from the microarray data of control individuals from the AD study in the input entities of the pathway model. Computational modeling and simulation of the inflammatory pathway enabled the comprehensive study of the system dynamics. Network and sensitivity analysis of the model unveiled essential interaction parameters and entities during neuroinflammation. Scanning of the key entities and parameters allowed us to determine their ultimate impact on neuronal apoptosis and survival. Moreover, the efficacy and potency of the FDA-approved drugs, namely Etanercept, Nivocasan, and Scyphostatin allowed us to study the model's response towards inhibition of the respective proteins/enzymes. The network analysis revealed the pivotal model entities with high betweenness and closeness centrality values including recruit FADD, TNFR_TRADD, act CASP2, actCASP8, actCASP3 and 9, cytochrome C, and RIP_RAIDD which profoundly impacted the neuronal apoptosis. Whereas some of the entities with high betweenness and closeness centrality values like Gi-coupled receptor, actS1PR, Sphingosine, S1P, actAKT, and actERK produced a high influence on neuronal survival. However, the current study inferred the dual role of ceramide, both on neuronal survival and apoptosis. Moreover, the drug Nivocasan effectively reduces neuronal apoptosis via its inhibitory mechanism on the caspases.
Collapse
Affiliation(s)
- Sanam Banaras
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Maryum Nisar
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Ayesha Arif
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Jamil Ahmad
- Computer Science and Information Technology (CS&IT), University of Malakand, Chakdara, Pakistan
| | - Muhammad Tariq Saeed
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Zartasha Mustansar
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | | | - Rizwan Nasir Paracha
- Department of Chemistry, University of Sargodha, Sub Campus Bhakkar, Bhakkar, Pakistan
| |
Collapse
|
26
|
Theiss EL, Griebsch LV, Lauer AA, Janitschke D, Erhardt VKJ, Haas EC, Kuppler KN, Radermacher J, Walzer O, Portius D, Grimm HS, Hartmann T, Grimm MOW. Vitamin B12 Attenuates Changes in Phospholipid Levels Related to Oxidative Stress in SH-SY5Y Cells. Cells 2022; 11:cells11162574. [PMID: 36010649 PMCID: PMC9406929 DOI: 10.3390/cells11162574] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/18/2022] [Accepted: 08/15/2022] [Indexed: 01/04/2023] Open
Abstract
Oxidative stress is closely linked to Alzheimer’s disease (AD), and is detected peripherally as well as in AD-vulnerable brain regions. Oxidative stress results from an imbalance between the generation and degradation of reactive oxidative species (ROS), leading to the oxidation of proteins, nucleic acids, and lipids. Extensive lipid changes have been found in post mortem AD brain tissue; these changes include the levels of total phospholipids, sphingomyelin, and ceramide, as well as plasmalogens, which are highly susceptible to oxidation because of their vinyl ether bond at the sn-1 position of the glycerol-backbone. Several lines of evidence indicate that a deficiency in the neurotropic vitamin B12 is linked with AD. In the present study, treatment of the neuroblastoma cell line SH-SY5Y with vitamin B12 resulted in elevated levels of phosphatidylcholine, phosphatidylethanolamine, sphingomyelin, and plasmalogens. Vitamin B12 also protected plasmalogens from hydrogen peroxide (H2O2)-induced oxidative stress due to an elevated expression of the ROS-degrading enzymes superoxide-dismutase (SOD) and catalase (CAT). Furthermore, vitamin B12 elevates plasmalogen synthesis by increasing the expression of alkylglycerone phosphate synthase (AGPS) and choline phosphotransferase 1 (CHPT1) in SH-SY5Y cells exposed to H2O2-induced oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Oliver Walzer
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
| | - Dorothea Portius
- Nutrition Therapy and Counseling, Campus Gera, SRH University of Applied Health Science, 07548 Gera, Germany
| | | | - Tobias Hartmann
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
- Deutsches Institut für DemenzPrävention, Saarland University, 66424 Homburg, Germany
| | - Marcus Otto Walter Grimm
- Experimental Neurology, Saarland University, 66424 Homburg, Germany
- Deutsches Institut für DemenzPrävention, Saarland University, 66424 Homburg, Germany
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, 51377 Leverkusen, Germany
- Correspondence: or
| |
Collapse
|
27
|
Winstone JK, Pathak KV, Winslow W, Piras IS, White J, Sharma R, Huentelman MJ, Pirrotte P, Velazquez R. Glyphosate infiltrates the brain and increases pro-inflammatory cytokine TNFα: implications for neurodegenerative disorders. J Neuroinflammation 2022; 19:193. [PMID: 35897073 PMCID: PMC9331154 DOI: 10.1186/s12974-022-02544-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/05/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Herbicides are environmental contaminants that have gained much attention due to the potential hazards they pose to human health. Glyphosate, the active ingredient in many commercial herbicides, is the most heavily applied herbicide worldwide. The recent rise in glyphosate application to corn and soy crops correlates positively with increased death rates due to Alzheimer's disease and other neurodegenerative disorders. Glyphosate has been shown to cross the blood-brain barrier in in vitro models, but has yet to be verified in vivo. Additionally, reports have shown that glyphosate exposure increases pro-inflammatory cytokines in blood plasma, particularly TNFα. METHODS Here, we examined whether glyphosate infiltrates the brain and elevates TNFα levels in 4-month-old C57BL/6J mice. Mice received either 125, 250, or 500 mg/kg/day of glyphosate, or a vehicle via oral gavage for 14 days. Urine, plasma, and brain samples were collected on the final day of dosing for analysis via UPLC-MS and ELISAs. Primary cortical neurons were derived from amyloidogenic APP/PS1 pups to evaluate in vitro changes in Aβ40-42 burden and cytotoxicity. RNA sequencing was performed on C57BL/6J brain samples to determine changes in the transcriptome. RESULTS Our analysis revealed that glyphosate infiltrated the brain in a dose-dependent manner and upregulated TNFα in both plasma and brain tissue post-exposure. Notably, glyphosate measures correlated positively with TNFα levels. Glyphosate exposure in APP/PS1 primary cortical neurons increases levels of soluble Aβ40-42 and cytotoxicity. RNAseq revealed over 200 differentially expressed genes in a dose-dependent manner and cell-type-specific deconvolution analysis showed enrichment of key biological processes in oligodendrocytes including myelination, axon ensheathment, glial cell development, and oligodendrocyte development. CONCLUSIONS Collectively, these results show for the first time that glyphosate infiltrates the brain, elevates both the expression of TNFα and soluble Aβ, and disrupts the transcriptome in a dose-dependent manner, suggesting that exposure to this herbicide may have detrimental outcomes regarding the health of the general population.
Collapse
Affiliation(s)
- Joanna K Winstone
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, 797 E Tyler St, Tempe, AZ, 85287, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Khyatiben V Pathak
- Integrated Mass Spectrometry Shared Resources (IMS-SR), City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Wendy Winslow
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, 797 E Tyler St, Tempe, AZ, 85287, USA
| | - Ignazio S Piras
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Jennifer White
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, 797 E Tyler St, Tempe, AZ, 85287, USA
| | - Ritin Sharma
- Integrated Mass Spectrometry Shared Resources (IMS-SR), City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Matthew J Huentelman
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Patrick Pirrotte
- Integrated Mass Spectrometry Shared Resources (IMS-SR), City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Ramon Velazquez
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, 797 E Tyler St, Tempe, AZ, 85287, USA.
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|
28
|
Zhao Y, Song P, Zhang H, Chen X, Han P, Yu X, Fang C, Xie F, Guo Q. Alteration of plasma metabolic profile and physical performance combined with metabolites is more sensitive to early screening for mild cognitive impairment. Front Aging Neurosci 2022; 14:951146. [PMID: 35959293 PMCID: PMC9360416 DOI: 10.3389/fnagi.2022.951146] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/29/2022] [Indexed: 11/25/2022] Open
Abstract
Objective Unbiased metabolic profiling has been initiated to identify novel metabolites. However, it remains a challenge to define reliable biomarkers for rapid and accurate diagnosis of mild cognitive impairment (MCI). Our study aimed to evaluate the association of serum metabolites with MCI, attempting to find new biomarkers and combination models that are distinct for MCI. Methods A total of 380 participants were recruited (mean age: 72.5 ± 5.19 years). We performed an untargeted metabolomics analysis on older adults who underwent the Mini-Mental State Examination (MMSE), the Instrumental Activities of Daily Living (IADL), and physical performance tests such as hand grip, Timed Up and Go Test (TUGT), and walking speed. Orthogonal partial least squares discriminant analysis (OPLS-DA) and heat map were utilized to distinguish the metabolites that differ between groups. Results Among all the subjects, 47 subjects were diagnosed with MCI, and methods based on the propensity score are used to match the MCI group with the normal control (NC) group (n = 47). The final analytic sample comprised 94 participants (mean age: 75.2 years). The data process from the metabolic profiles identified 1,008 metabolites. A cluster and pathway enrichment analysis showed that sphingolipid metabolism is involved in the development of MCI. Combination of metabolite panel and physical performance were significantly increased discriminating abilities on MCI than a single physical performance test [model 1: the area under the curve (AUC) = 0.863; model 2: AUC = 0.886; and model 3: AUC = 0.870, P < 0.001]. Conclusion In our study, untargeted metabolomics was used to detect the disturbance of metabolism that occurs in MCI. Physical performance tests combined with phosphatidylcholines (PCs) showed good utility in discriminating between NC and MCI, which is meaningful for the early diagnosis of MCI.
Collapse
Affiliation(s)
- Yinjiao Zhao
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Peiyu Song
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Hui Zhang
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Xiaoyu Chen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Peipei Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xing Yu
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chenghu Fang
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Fandi Xie
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Qi Guo
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Qi Guo
| |
Collapse
|
29
|
Zhu Z, Quadri Z, Crivelli SM, Elsherbini A, Zhang L, Tripathi P, Qin H, Roush E, Spassieva SD, Nikolova-Karakashian M, McClintock TS, Bieberich E. Neutral Sphingomyelinase 2 Mediates Oxidative Stress Effects on Astrocyte Senescence and Synaptic Plasticity Transcripts. Mol Neurobiol 2022; 59:3233-3253. [PMID: 35294731 PMCID: PMC9023069 DOI: 10.1007/s12035-022-02747-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 01/11/2022] [Indexed: 11/26/2022]
Abstract
We have shown that deficiency of neutral sphingomyelinase 2 (nSMase2), an enzyme generating the sphingolipid ceramide, improves memory in adult mice. Here, we performed sphingolipid and RNA-seq analyses on the cortex from 10-month-old nSMase2-deficient (fro/fro) and heterozygous (+ /fro) mice. fro/fro cortex showed reduced levels of ceramide, particularly in astrocytes. Differentially abundant transcripts included several functionally related groups, with decreases in mitochondrial oxidative phosphorylation and astrocyte activation transcripts, while axon guidance and synaptic transmission and plasticity transcripts were increased, indicating a role of nSMase2 in oxidative stress, astrocyte activation, and cognition. Experimentally induced oxidative stress decreased the level of glutathione (GSH), an endogenous inhibitor of nSMase2, and increased immunolabeling for ceramide in primary + /fro astrocytes, but not in fro/fro astrocytes. β-galactosidase activity was lower in 5-week-old fro/fro astrocytes, indicating delayed senescence due to nSMase2 deficiency. In fro/fro cortex, levels of the senescence markers C3b and p27 and the proinflammatory cytokines interleukin 1β, interleukin 6, and tumor necrosis factor α were reduced, concurrent with twofold decreased phosphorylation of their downstream target, protein kinase Stat3. RNA and protein levels of the ionotropic glutamate receptor subunit 2B (Grin2b/NR2B) were increased by twofold, which was previously shown to enhance cognition. This was consistent with threefold reduced levels of exosomes carrying miR-223-3p, a micro-RNA downregulating NR2B. In summary, our data show that nSMase2 deficiency prevents oxidative stress-induced elevation of ceramide and secretion of exosomes by astrocytes that suppress neuronal function, indicating a role of nSMase2 in the regulation of neuroinflammation and cognition.
Collapse
Affiliation(s)
- Zhihui Zhu
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Zainuddin Quadri
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Simone M. Crivelli
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Ahmed Elsherbini
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Liping Zhang
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Priyanka Tripathi
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Haiyan Qin
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Emily Roush
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Stefka D. Spassieva
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | | | - Timothy S. McClintock
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Erhard Bieberich
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| |
Collapse
|
30
|
Liu Y, Zhang X, Lin W, Kehriman N, Kuang W, Ling X. Multi-factor combined biomarker screening strategy to rapidly diagnose Alzheimer’s disease and evaluate drug effect based on a rat model. J Pharm Anal 2022; 12:627-636. [PMID: 36105160 PMCID: PMC9463486 DOI: 10.1016/j.jpha.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/04/2022] Open
Abstract
Alzheimer's disease (AD) represents the main form of dementia; however, valid diagnosis and treatment measures are lacking. The discovery of valuable biomarkers through omics technologies can help solve this problem. For this reason, metabolomic analysis using ultra-performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS) was carried out on plasma, hippocampus, and cortex samples of an AD rat model. Based on the metabolomic data, we report a multi-factor combined biomarker screening strategy to rapidly and accurately identify potential biomarkers. Compared with the usual procedure, our strategy can identify fewer biomarkers with higher diagnostic specificity and sensitivity. In addition to diagnosis, the potential biomarkers identified using our strategy were also beneficial for drug evaluation. Multi-factor combined biomarker screening strategy was used to identify differential metabolites from a rat model of amyloid beta peptide 1–40 (Aβ1−40) plus ibotenic acid-induced AD (compared with the controls) for the first time; lysophosphatidylcholine (LysoPC) and intermediates of sphingolipid metabolism were screened as potential biomarkers. Subsequently, the effects of donepezil and pine nut were successfully reflected by regulating the levels of the abovementioned biomarkers and metabolic profile distribution in partial least squares-discriminant analysis (PLS-DA). This novel biomarker screening strategy can be used to analyze other metabolomic data to simultaneously enable disease diagnosis and drug evaluation. Multi-factor combined biomarker screening strategy is a novel and rapid metabolomic data processing strategy. The most discriminating biomarkers for AD diagnosis can simultaneously reflect drug effects. Multi-factor biomarker screening strategy is ready for use without a priori knowledge.
Collapse
|
31
|
Solomon V, Hafez M, Xian H, Harrington M, Fonteh A, Yassine H. An Association Between Saturated Fatty Acid-Containing Phosphatidylcholine in Cerebrospinal Fluid with Tau Phosphorylation. J Alzheimers Dis 2022; 87:609-617. [DOI: 10.3233/jad-215643] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Mechanistic studies in animal models implicate a role for saturated fatty acids in neurodegeneration, but validation of this finding in human studies is still lacking. Objective: We investigated how cerebrospinal levels of sphingomyelins (SM) and phosphatidylcholine (PC)-containing saturated fatty acids, monounsaturated fatty acids, and polyunsaturated fatty acids associate with total tau and phosphorylated tau (p-tau). Methods: Cerebrospinal fluid (CSF) lipids were measured in two cohorts, a discovery and a confirmation cohort of older non-demented individuals from University of Southern California and Huntington Medical Research Institutes cohorts. Lipid analysis was performed using hydrophilic interaction liquid chromatography, and individual PC and SM lipid species were measured using tandem mass spectrometry. In addition, CSF levels of Aβ 42, total tau, and p-tau-181 were measured using an MSD multiplex assay. Results: The discovery cohort (n = 47) consisted of older individuals and more females compared to the confirmation cohort (n = 46). Notwithstanding the age and gender differences, and a higher p-tau, Aβ 42, and LDL-cholesterol in the discovery cohort, CSF concentrations of dipalmitoyl-PC (PC32a:0) were significantly associated with p-tau in both cohorts. Similarly, total saturated PC but not mono or polyunsaturated PCs correlated with p-tau concentrations in both cohorts. Conclusion: Saturated PC species in CSF associate with early markers of neurodegeneration and are potential early disease progression biomarkers. We propose mechanisms by which saturated PC may promote tau hyperphosphorylation.
Collapse
Affiliation(s)
- Victoria Solomon
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Madonna Hafez
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Haotian Xian
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Michael Harrington
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Alfred Fonteh
- Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, USA
| | - Hussein Yassine
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
32
|
Loft LMI, Moseholm KF, Pedersen KKW, Jensen MK, Koch M, Cronjé HT. Sphingomyelins and ceramides: possible biomarkers for dementia? Curr Opin Lipidol 2022; 33:57-67. [PMID: 34879042 DOI: 10.1097/mol.0000000000000804] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW Dementia is a public health challenge with no existing cure or early biomarkers. We review the evidence for blood-based measures of sphingomyelins and ceramides as potential novel biomarkers of dementia. RECENT FINDINGS In recent years, lipids have been under investigation for their role in neurodegenerative diseases especially dementia and Alzheimer's disease. Increasing evidence from postmortem human brains suggests that alterations in the metabolism of sphingolipids could play a crucial part in dementia. Findings from epidemiological investigations of blood-based sphingomyelins and ceramides have been inconsistent. SUMMARY This review focuses on blood-based measures of 10 specific ceramides and sphingomyelins (Cer C16:0, Cer C20:0, Cer C22:0, Cer C24:0, Cer C24:1 and SM C16:0, SM C20:0, SM C22:0, SM C24:0, SM C24:1) in relation to cognition and dementia. On the bais of 15 studies, there was no robust association between ceramide and sphingomyelin levels and prevalent or incident dementia. Cross-sectionally, Cer C16:0 and Cer C24:1 tends to be higher in dementia cases vs. controls.
Collapse
Affiliation(s)
- Laura M I Loft
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| | - Kristine F Moseholm
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| | - Karoline K W Pedersen
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| | - Majken K Jensen
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
- Harvard T.H. Chan School of Public Health, Department of Nutrition, Boston, Massachusetts, USA
| | - Manja Koch
- Harvard T.H. Chan School of Public Health, Department of Nutrition, Boston, Massachusetts, USA
- Institute of Epidemiology University Hospital Schleswig-Holstein, University of Kiel, Kiel, Germany
| | - Héléne T Cronjé
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Thomas JM, Sudhadevi T, Basa P, Ha AW, Natarajan V, Harijith A. The Role of Sphingolipid Signaling in Oxidative Lung Injury and Pathogenesis of Bronchopulmonary Dysplasia. Int J Mol Sci 2022; 23:ijms23031254. [PMID: 35163176 PMCID: PMC8835774 DOI: 10.3390/ijms23031254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Premature infants are born with developing lungs burdened by surfactant deficiency and a dearth of antioxidant defense systems. Survival rate of such infants has significantly improved due to advances in care involving mechanical ventilation and oxygen supplementation. However, a significant subset of such survivors develops the chronic lung disease, Bronchopulmonary dysplasia (BPD), characterized by enlarged, simplified alveoli and deformed airways. Among a host of factors contributing to the pathogenesis is oxidative damage induced by exposure of the developing lungs to hyperoxia. Recent data indicate that hyperoxia induces aberrant sphingolipid signaling, leading to mitochondrial dysfunction and abnormal reactive oxygen species (ROS) formation (ROS). The role of sphingolipids such as ceramides and sphingosine 1-phosphate (S1P), in the development of BPD emerged in the last decade. Both ceramide and S1P are elevated in tracheal aspirates of premature infants of <32 weeks gestational age developing BPD. This was faithfully reflected in the murine models of hyperoxia and BPD, where there is an increased expression of sphingolipid metabolites both in lung tissue and bronchoalveolar lavage. Treatment of neonatal pups with a sphingosine kinase1 specific inhibitor, PF543, resulted in protection against BPD as neonates, accompanied by improved lung function and reduced airway remodeling as adults. This was accompanied by reduced mitochondrial ROS formation. S1P receptor1 induced by hyperoxia also aggravates BPD, revealing another potential druggable target in this pathway for BPD. In this review we aim to provide a detailed description on the role played by sphingolipid signaling in hyperoxia induced lung injury and BPD.
Collapse
Affiliation(s)
- Jaya M. Thomas
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Prathima Basa
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
| | - Alison W. Ha
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (J.M.T.); (T.S.); (P.B.); (A.W.H.)
- Correspondence: ; Tel.: +1-(216)-286-7038
| |
Collapse
|
34
|
Quintero ME, Pontes JGDM, Tasic L. Metabolomics in degenerative brain diseases. Brain Res 2021; 1773:147704. [PMID: 34744014 DOI: 10.1016/j.brainres.2021.147704] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/18/2021] [Accepted: 10/23/2021] [Indexed: 12/23/2022]
Abstract
Among the most studied diseases that affect the central nervous system are Parkinson's, Alzheimer's, and Huntington's diseases, but the lack of effective biomarkers, accurate diagnosis, and precise treatment for each of them is currently an issue. Due to the contribution of biomarkers in supporting diagnosis, many recent efforts have focused on their identification and validation at the beginning or during the progression of the mental illness. Metabolome reveals the metabolic processes that result from protein activities under the guided gene expression and environmental factors, either in healthy or pathological conditions. In this context, metabolomics has proven to be a valuable approach. Currently, magnetic resonance spectroscopy (NMR) and mass spectrometry (MS) are the most commonly used bioanalytical techniques for metabolomics. MS-assisted profiling is considered the most versatile technique, and the NMR is the most reproductive. However, each one of them has its drawbacks. In this review, we summarized several alterations in metabolites that have been reported for these three classic brain diseases using MS and NMR-based research, which might suggest some possible biomarkers to support the diagnosis and/or new targets for their treatment.
Collapse
Affiliation(s)
- Melissa Escobar Quintero
- Laboratory of Chemical Biology, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - João Guilherme de Moraes Pontes
- Laboratory of Chemical Biology, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ljubica Tasic
- Laboratory of Chemical Biology, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
35
|
Marini M, Tani A, Manetti M, Sgambati E. Overview of sialylation status in human nervous and skeletal muscle tissues during aging. Acta Histochem 2021; 123:151813. [PMID: 34753032 DOI: 10.1016/j.acthis.2021.151813] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022]
Abstract
Sialic acids (Sias) are a large and heterogeneous family of electronegatively charged nine-carbon monosaccharides containing a carboxylic acid and are mostly found as terminal residues in glycans of glycoproteins and glycolipids such as gangliosides. They are linked to galactose or N-acetylgalactosamine via α2,3 or α2,6 linkage, or to other Sias via α2,8 or more rarely α2,9 linkage, resulting in mono, oligo and polymeric forms. Given their characteristics, Sias play a crucial role in a multitude of human tissue biological processes in physiological and pathological conditions, ranging from development and growth to adult life until aging. Here, we review the sialylation status in human adult life focusing on the nervous and skeletal muscle tissues, which both display significant structural and functional changes during aging, strongly impacting on the whole human body and, therefore, on the quality of life. In particular, this review highlights the fundamental roles played by different types of glycoconjugates Sias in several cellular biological processes in the nervous and skeletal muscle tissues during adult life, also discussing how changes in Sia content during aging may contribute to the physiological decline of physical and nervous functions and to the development of age-related degenerative pathologies. Based on our current knowledge, further in-depth investigations could help to develop novel prophylactic strategies and therapeutic approaches that, by maintaining and/or restoring the correct sialylation status in the nervous and skeletal muscle tissues, could contribute to aging slowing and the prevention of age-related pathologies.
Collapse
|
36
|
Lipids in Pathophysiology and Development of the Membrane Lipid Therapy: New Bioactive Lipids. MEMBRANES 2021; 11:membranes11120919. [PMID: 34940418 PMCID: PMC8708953 DOI: 10.3390/membranes11120919] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Membranes are mainly composed of a lipid bilayer and proteins, constituting a checkpoint for the entry and passage of signals and other molecules. Their composition can be modulated by diet, pathophysiological processes, and nutritional/pharmaceutical interventions. In addition to their use as an energy source, lipids have important structural and functional roles, e.g., fatty acyl moieties in phospholipids have distinct impacts on human health depending on their saturation, carbon length, and isometry. These and other membrane lipids have quite specific effects on the lipid bilayer structure, which regulates the interaction with signaling proteins. Alterations to lipids have been associated with important diseases, and, consequently, normalization of these alterations or regulatory interventions that control membrane lipid composition have therapeutic potential. This approach, termed membrane lipid therapy or membrane lipid replacement, has emerged as a novel technology platform for nutraceutical interventions and drug discovery. Several clinical trials and therapeutic products have validated this technology based on the understanding of membrane structure and function. The present review analyzes the molecular basis of this innovative approach, describing how membrane lipid composition and structure affects protein-lipid interactions, cell signaling, disease, and therapy (e.g., fatigue and cardiovascular, neurodegenerative, tumor, infectious diseases).
Collapse
|
37
|
den Hoedt S, Crivelli SM, Leijten FPJ, Losen M, Stevens JAA, Mané-Damas M, de Vries HE, Walter J, Mirzaian M, Sijbrands EJG, Aerts JMFG, Verhoeven AJM, Martinez-Martinez P, Mulder MT. Effects of Sex, Age, and Apolipoprotein E Genotype on Brain Ceramides and Sphingosine-1-Phosphate in Alzheimer's Disease and Control Mice. Front Aging Neurosci 2021; 13:765252. [PMID: 34776936 PMCID: PMC8579780 DOI: 10.3389/fnagi.2021.765252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/29/2021] [Indexed: 11/28/2022] Open
Abstract
Apolipoprotein ε4 (APOE)4 is a strong risk factor for the development of Alzheimer’s disease (AD) and aberrant sphingolipid levels have been implicated in AD. We tested the hypothesis that the APOE4 genotype affects brain sphingolipid levels in AD. Seven ceramides and sphingosine-1-phosphate (S1P) were quantified by LC-MSMS in hippocampus, cortex, cerebellum, and plasma of <3 months and >5 months old human APOE3 and APOE4-targeted replacement mice with or without the familial AD (FAD) background of both sexes (145 animals). APOE4 mice had higher Cer(d18:1/24:0) levels in the cortex (1.7-fold, p = 0.002) than APOE3 mice. Mice with AD background showed higher levels of Cer(d18:1/24:1) in the cortex than mice without (1.4-fold, p = 0.003). S1P levels were higher in all three brain regions of older mice than of young mice (1.7-1.8-fold, all p ≤ 0.001). In female mice, S1P levels in hippocampus (r = −0.54 [−0.70, −0.35], p < 0.001) and in cortex correlated with those in plasma (r = −0.53 [−0.71, −0.32], p < 0.001). Ceramide levels were lower in the hippocampus (3.7–10.7-fold, all p < 0.001), but higher in the cortex (2.3–12.8-fold, p < 0.001) of female than male mice. In cerebellum and plasma, sex effects on individual ceramides depended on acyl chain length (9.5-fold lower to 11.5-fold higher, p ≤ 0.001). In conclusion, sex is a stronger determinant of brain ceramide levels in mice than APOE genotype, AD background, or age. Whether these differences impact AD neuropathology in men and women remains to be investigated.
Collapse
Affiliation(s)
- Sandra den Hoedt
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Simone M Crivelli
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Frank P J Leijten
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Mario Losen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Jo A A Stevens
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Marina Mané-Damas
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, VU Medical Center, Amsterdam UMC, Amsterdam, Netherlands
| | - Jochen Walter
- Department of Neurology, University Hospital Bonn, Venusberg Campus, Bonn, Germany
| | - Mina Mirzaian
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Adrie J M Verhoeven
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pilar Martinez-Martinez
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
38
|
Bu Y, Wu H, Deng R, Wang Y. Therapeutic Potential of SphK1 Inhibitors Based on Abnormal Expression of SphK1 in Inflammatory Immune Related-Diseases. Front Pharmacol 2021; 12:733387. [PMID: 34737701 PMCID: PMC8560647 DOI: 10.3389/fphar.2021.733387] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/04/2021] [Indexed: 01/12/2023] Open
Abstract
Sphingosine kinase 1(SphK1) a key enzyme that catalyzes the conversion of sphingosine (Sph) to sphingosine 1-phosphate (S1P), so as to maintain the dynamic balance of sphingolipid-rheostat in cells and participate in cell growth and death, proliferation and migration, vasoconstriction and remodeling, inflammation and metabolism. The normal expression of SphK1 maintains the balance of physiological and pathological states, which is reflected in the regulation of inflammatory factor secretion, immune response in traditional immune cells and non-traditional immune cells, and complex signal transduction. However, abnormal SphK1 expression and activity are found in various inflammatory and immune related-diseases, such as hypertension, atherosclerosis, Alzheimer’s disease, inflammatory bowel disease and rheumatoid arthritis. In view of the therapeutic potential of regulating SphK1 and its signal, the current research is aimed at SphK1 inhibitors, such as SphK1 selective inhibitors and dual SphK1/2 inhibitor, and other compounds with inhibitory potency. This review explores the regulatory role of over-expressed SphK1 in inflammatory and immune related-diseases, and investigate the latest progress of SphK1 inhibitors and the improvement of disease or pathological state.
Collapse
Affiliation(s)
- Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.,Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| |
Collapse
|
39
|
The Hitchhiker's Guide to Untargeted Lipidomics Analysis: Practical Guidelines. Metabolites 2021; 11:metabo11110713. [PMID: 34822371 PMCID: PMC8624948 DOI: 10.3390/metabo11110713] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 11/30/2022] Open
Abstract
Lipidomics is a newly emerged discipline involving the identification and quantification of thousands of lipids. As a part of the omics field, lipidomics has shown rapid growth both in the number of studies and in the size of lipidome datasets, thus, requiring specific and efficient data analysis approaches. This paper aims to provide guidelines for analyzing and interpreting lipidome data obtained using untargeted methods that rely on liquid chromatography coupled with mass spectrometry (LC-MS) to detect and measure the intensities of lipid compounds. We present a state-of-the-art untargeted LC-MS workflow for lipidomics, from study design to annotation of lipid features, focusing on practical, rather than theoretical, approaches for data analysis, and we outline possible applications of untargeted lipidomics for biological studies. We provide a detailed R notebook designed specifically for untargeted lipidome LC-MS data analysis, which is based on xcms software.
Collapse
|
40
|
Johnson JO, Chia R, Miller DE, Li R, Kumaran R, Abramzon Y, Alahmady N, Renton AE, Topp SD, Gibbs JR, Cookson MR, Sabir MS, Dalgard CL, Troakes C, Jones AR, Shatunov A, Iacoangeli A, Al Khleifat A, Ticozzi N, Silani V, Gellera C, Blair IP, Dobson-Stone C, Kwok JB, Bonkowski ES, Palvadeau R, Tienari PJ, Morrison KE, Shaw PJ, Al-Chalabi A, Brown RH, Calvo A, Mora G, Al-Saif H, Gotkine M, Leigh F, Chang IJ, Perlman SJ, Glass I, Scott AI, Shaw CE, Basak AN, Landers JE, Chiò A, Crawford TO, Smith BN, Traynor BJ. Association of Variants in the SPTLC1 Gene With Juvenile Amyotrophic Lateral Sclerosis. JAMA Neurol 2021; 78:1236-1248. [PMID: 34459874 PMCID: PMC8406220 DOI: 10.1001/jamaneurol.2021.2598] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
Importance Juvenile amyotrophic lateral sclerosis (ALS) is a rare form of ALS characterized by age of symptom onset less than 25 years and a variable presentation. Objective To identify the genetic variants associated with juvenile ALS. Design, Setting, and Participants In this multicenter family-based genetic study, trio whole-exome sequencing was performed to identify the disease-associated gene in a case series of unrelated patients diagnosed with juvenile ALS and severe growth retardation. The patients and their family members were enrolled at academic hospitals and a government research facility between March 1, 2016, and March 13, 2020, and were observed until October 1, 2020. Whole-exome sequencing was also performed in a series of patients with juvenile ALS. A total of 66 patients with juvenile ALS and 6258 adult patients with ALS participated in the study. Patients were selected for the study based on their diagnosis, and all eligible participants were enrolled in the study. None of the participants had a family history of neurological disorders, suggesting de novo variants as the underlying genetic mechanism. Main Outcomes and Measures De novo variants present only in the index case and not in unaffected family members. Results Trio whole-exome sequencing was performed in 3 patients diagnosed with juvenile ALS and their parents. An additional 63 patients with juvenile ALS and 6258 adult patients with ALS were subsequently screened for variants in the SPTLC1 gene. De novo variants in SPTLC1 (p.Ala20Ser in 2 patients and p.Ser331Tyr in 1 patient) were identified in 3 unrelated patients diagnosed with juvenile ALS and failure to thrive. A fourth variant (p.Leu39del) was identified in a patient with juvenile ALS where parental DNA was unavailable. Variants in this gene have been previously shown to be associated with autosomal-dominant hereditary sensory autonomic neuropathy, type 1A, by disrupting an essential enzyme complex in the sphingolipid synthesis pathway. Conclusions and Relevance These data broaden the phenotype associated with SPTLC1 and suggest that patients presenting with juvenile ALS should be screened for variants in this gene.
Collapse
Affiliation(s)
- Janel O. Johnson
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Ruth Chia
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Danny E. Miller
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle
- Department of Pediatrics, Division of Genetic Medicine, Seattle Children’s Hospital, University of Washington, Seattle
| | - Rachel Li
- Department of Pediatrics, Children’s Hospital of Richmond at VCU, Richmond, Virginia
| | - Ravindran Kumaran
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Yevgeniya Abramzon
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Nada Alahmady
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Biology, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Alan E. Renton
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Simon D. Topp
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- UK Dementia Research Institute at King’s College London, London, United Kingdom
| | - J. Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Mark R. Cookson
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Marya S. Sabir
- Neurodegenerative Diseases Research Unit, Laboratory of Neurogenetics, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- The American Genome Center, Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Claire Troakes
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Ashley R. Jones
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Nicola Ticozzi
- Istituto Auxologico Italiano, IRCCS, Department of Neurology–Stroke Unit and Laboratory of Neuroscience, Milan, Italy
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan, Italy
| | - Vincenzo Silani
- Istituto Auxologico Italiano, IRCCS, Department of Neurology–Stroke Unit and Laboratory of Neuroscience, Milan, Italy
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan, Italy
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico ‘Carlo Besta,’ Milan, Italy
| | - Ian P. Blair
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Carol Dobson-Stone
- The University of Sydney, Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, Camperdown, Australia
- School of Medical Sciences, University of New South Wales, Kensington, Australia
| | - John B. Kwok
- The University of Sydney, Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, Camperdown, Australia
- School of Medical Sciences, University of New South Wales, Kensington, Australia
| | - Emily S. Bonkowski
- Department of Pediatrics, Division of Genetic Medicine, Seattle Children’s Hospital, University of Washington, Seattle
| | - Robin Palvadeau
- Suna and Inan Kırac Foundation, Neurodegeneration Research Laboratory, KUTTAM, School of Medicine, Koc University, Istanbul, Turkey
| | - Pentti J. Tienari
- Department of Neurology, Helsinki University Hospital and Translational Immunology Programme, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Karen E. Morrison
- Faculty of Medicine, Health and Life Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Neurology, King’s College Hospital, London, United Kingdom
| | - Robert H. Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester
| | - Andrea Calvo
- ALS Center, ‘Rita Levi Montalcini’ Department of Neuroscience, University of Turin, Turin, Italy
| | | | - Hind Al-Saif
- Department of Neurology, Children’s Hospital of Richmond at VCU, Richmond, Virginia
| | - Marc Gotkine
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Fawn Leigh
- Department of Neurology, Seattle Children’s Hospital, University of Washington, Seattle
| | - Irene J. Chang
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle
| | - Seth J. Perlman
- Department of Neurology, Seattle Children’s Hospital, University of Washington, Seattle
| | - Ian Glass
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle
- Department of Pediatrics, Division of Genetic Medicine, Seattle Children’s Hospital, University of Washington, Seattle
| | - Anna I. Scott
- Department of Laboratories, Seattle Children’s Hospital, Seattle, Washington
- Department of Laboratory Medicine, University of Washington, Seattle
| | - Christopher E. Shaw
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- UK Dementia Research Institute at King’s College London, London, United Kingdom
| | - A. Nazli Basak
- Suna and Inan Kırac Foundation, Neurodegeneration Research Laboratory, KUTTAM, School of Medicine, Koc University, Istanbul, Turkey
| | - John E. Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester
| | - Adriano Chiò
- ALS Center, ‘Rita Levi Montalcini’ Department of Neuroscience, University of Turin, Turin, Italy
- Neurology 1, AOU Città della Salute e della Scienza, Turin, Italy
| | - Thomas O. Crawford
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland
| | - Bradley N. Smith
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Bryan J. Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | | |
Collapse
|
41
|
Sriwichaiin S, Chattipakorn N, Chattipakorn SC. Metabolomic Alterations in the Blood and Brain in Association with Alzheimer's Disease: Evidence from in vivo to Clinical Studies. J Alzheimers Dis 2021; 84:23-50. [PMID: 34511504 DOI: 10.3233/jad-210737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) has become a major health problem among the elderly population. Some evidence suggests that metabolic disturbance possibly plays a role in the pathophysiology of AD. Currently, the study of metabolomics has been used to explore changes in multiple metabolites in several diseases, including AD. Thus, the metabolomics research in AD might provide some information regarding metabolic dysregulations, and their possible associated pathophysiology. This review summarizes the information discovered regarding the metabolites in the brain and the blood from the metabolomics research of AD from both animal and clinical studies. Additionally, the correlation between the changes in metabolites and outcomes, such as pathological findings in the brain and cognitive impairment are discussed. We also deliberate on the findings of cohort studies, demonstrating the alterations in metabolites before changes of cognitive function. All of these findings can be used to inform the potential identity of specific metabolites as possible biomarkers for AD.
Collapse
Affiliation(s)
- Sirawit Sriwichaiin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
42
|
Barron K, Ogretmen B, Krupenko N. Ceramide synthase 6 mediates sex-specific metabolic response to dietary folic acid in mice. J Nutr Biochem 2021; 98:108832. [PMID: 34358645 DOI: 10.1016/j.jnutbio.2021.108832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/20/2021] [Accepted: 07/01/2021] [Indexed: 11/26/2022]
Abstract
Folic acid-fortified foods and multi-vitamin supplements containing folic acid (FA) are widely used around the world, but the exact mechanisms/metabolic effects of FA are not precisely identified. We have demonstrated that Ceramide Synthase 6 (CerS6) and C16:0-ceramide mediate response to folate stress in cultured cells. Here we investigated the dietary FA effects on mouse liver metabolome, with a specific focus on sphingolipids, CerS6 and C16:0-ceramide. Wild-type and CerS6-/- mice were fed FA-deficient, control, or FA over-supplemented diets for 4 weeks. After dietary treatment, liver concentrations of ceramides, sphingomyelins and hexosylceramides were measured by LC-MS/MS and complemented by untargeted metabolomic characterization of mouse livers. Our study shows that alterations in dietary FA elicit multiple sphingolipid responses mediated by CerS6 in mouse livers. Folic acid-deficient diet elevated C14:0-, C18:0- and C20:0- but not C16:0-ceramide in WT male and female mice. Additionally, FA over-supplementation increased multiple sphingomyelin species, including total sphingomyelins, in both sexes. Of note, concentrations of C14:0- and C16:0-ceramides and hexosylceramides were significantly higher in female livers than in male. The latter were increased by FD diet, with no difference between sexes in total pools of these sphingolipid classes. Untargeted liver metabolomic analysis concurred with the targeted measurements and showed broad effects of dietary FA and CerS6 status on multiple lipid classes including sex-specific effects on phosphatidylethanolamines and diacylglycerols. Our study demonstrates that both dietary FA and CerS6 status exhibit pleiotropic and sex-dependent effects on liver metabolism, including hepatic sphingolipids, diacylglycerols, long chain fatty acids, and phospholipids.
Collapse
Affiliation(s)
- Keri Barron
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Besim Ogretmen
- Department of Biochemistry & Molecular Biology, Hollings Cancer center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, South Carolina
| | - Natalia Krupenko
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina,; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina,.
| |
Collapse
|
43
|
Nho K, Kueider-Paisley A, Arnold M, MahmoudianDehkordi S, Risacher SL, Louie G, Blach C, Baillie R, Han X, Kastenmüller G, Doraiswamy PM, Kaddurah-Daouk R, Saykin AJ. Serum metabolites associated with brain amyloid beta deposition, cognition and dementia progression. Brain Commun 2021; 3:fcab139. [PMID: 34396103 PMCID: PMC8361396 DOI: 10.1093/braincomms/fcab139] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/30/2021] [Indexed: 11/13/2022] Open
Abstract
Metabolomics in the Alzheimer's Disease Neuroimaging Initiative cohort provides a powerful tool for mapping biochemical changes in Alzheimer's disease, and a unique opportunity to learn about the association between circulating blood metabolites and brain amyloid-β deposition in Alzheimer's disease. We examined 140 serum metabolites and their associations with brain amyloid-β deposition, cognition and conversion from mild cognitive impairment to Alzheimer's disease in the Alzheimer's Disease Neuroimaging Initiative. Processed [18F] Florbetapir PET images were used to perform a voxel-wise statistical analysis of the effect of metabolite levels on amyloid-β accumulation across the whole brain. We performed a multivariable regression analysis using age, sex, body mass index, apolipoprotein E ε4 status and study phase as covariates. We identified nine metabolites as significantly associated with amyloid-β deposition after multiple comparison correction. Higher levels of one acylcarnitine (C3; propionylcarnitine) and one biogenic amine (kynurenine) were associated with decreased amyloid-β accumulation and higher memory scores. However, higher levels of seven phosphatidylcholines (lysoPC a C18:2, PC aa C42:0, PC ae C42:3, PC ae C44:3, PC ae C44:4, PC ae C44:5 and PC ae C44:6) were associated with increased brain amyloid-β deposition. In addition, higher levels of PC ae C44:4 were significantly associated with lower memory and executive function scores and conversion from mild cognitive impairment to Alzheimer's disease dementia. Our findings suggest that dysregulation of peripheral phosphatidylcholine metabolism is associated with earlier pathological changes noted in Alzheimer's disease as measured by brain amyloid-β deposition as well as later clinical features including changes in memory and executive functioning. Perturbations in phosphatidylcholine metabolism may point to issues with membrane restructuring leading to the accumulation of amyloid-β in the brain. Additional studies are needed to explore whether these metabolites play a causal role in the pathogenesis of Alzheimer's disease or if they are biomarkers for systemic changes during preclinical phases of the disease.
Collapse
Affiliation(s)
- Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27710, USA
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | | | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gregory Louie
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27710, USA
| | - Colette Blach
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | | | - Xianlin Han
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78249, USA
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27710, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC 27710, USA
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27710, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC 27710, USA
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
44
|
Korshoj LE, Kielian T. Neuroimmune metabolism: Uncovering the role of metabolic reprogramming in central nervous system disease. J Neurochem 2021; 158:8-13. [PMID: 33993505 DOI: 10.1111/jnc.15376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/16/2022]
Abstract
The rapidly expanding field of immunometabolism has highlighted an intricate association between the metabolic pathways that program cellular pro-inflammatory versus anti-inflammatory activity. This Special Issue on Neuroimmune Metabolism showcases a growing body of work characterizing the metabolic profiles of the major CNS-resident and peripheral immune cell players in neuroinflammation, neurodegeneration, and brain injury. The review articles address the roles of glycolytic, oxidative, and lipid metabolism that are associated with beneficial or detrimental properties in various neurological conditions, address unanswered questions in the field, and discuss promising avenues for future therapeutics. Cover Image for this issue: https://doi.org/10.1111/jnc.15069.
Collapse
Affiliation(s)
- Lee E Korshoj
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
45
|
Teitsdottir UD, Halldorsson S, Rolfsson O, Lund SH, Jonsdottir MK, Snaedal J, Petersen PH. Cerebrospinal Fluid C18 Ceramide Associates with Markers of Alzheimer's Disease and Inflammation at the Pre- and Early Stages of Dementia. J Alzheimers Dis 2021; 81:231-244. [PMID: 33814423 PMCID: PMC8203241 DOI: 10.3233/jad-200964] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Understanding how dysregulation in lipid metabolism relates to the severity of Alzheimer‘s disease (AD) pathology might be critical in developing effective treatments. Objective: To identify lipid species in cerebrospinal fluid (CSF) associated with signature AD pathology and to explore their relationships with measures reflecting AD-related processes (neurodegeneration, inflammation, deficits in verbal episodic memory) among subjects at the pre- and early symptomatic stages of dementia. Methods: A total of 60 subjects that had been referred to an Icelandic memory clinic cohort were classified as having CSF AD (n = 34) or non-AD (n = 26) pathology profiles. Untargeted CSF lipidomic analysis was performed using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS) for the detection of mass-to-charge ratio (m/z) features. CSF proteins reflecting neurodegeneration (neurofilament light [NFL]) and inflammation (chitinase-3-like protein 1 [YKL-40], S100 calcium-binding protein B [S100B], glial fibrillary acidic protein [GFAP]) were also measured. Rey Auditory Verbal Learning (RAVLT) and Story tests were used for the assessment of verbal episodic memory. Results: Eight out of 1008 features were identified as best distinguishing between the CSF profile groups. Of those, only the annotation of the m/z feature assigned to lipid species C18 ceramide was confirmed with a high confidence. Multiple regression analyses, adjusted for age, gender, and education, demonstrated significant associations of CSF core AD markers (Aβ42: st.β= –0.36, p = 0.007; T-tau: st.β= 0.41, p = 0.005) and inflammatory marker S100B (st.β= 0.51, p = 0.001) with C18 ceramide levels. Conclusion: Higher levels of C18 ceramide associated with increased AD pathology and inflammation, suggesting its potential value as a therapeutic target.
Collapse
Affiliation(s)
- Unnur D Teitsdottir
- Faculty of Medicine, Department of Anatomy, Biomedical Center, University of Iceland, Reykjavik, Iceland
| | | | - Ottar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
| | | | - Maria K Jonsdottir
- Department of Psychology, Reykjavik University, Reykjavik, Iceland.,Department of Psychiatry, Landspitali -National University Hospital, Reykjavik, Iceland
| | - Jon Snaedal
- Memory Clinic, Department of Geriatric Medicine, Landspitali - National University Hospital, Reykjavik, Iceland
| | - Petur H Petersen
- Faculty of Medicine, Department of Anatomy, Biomedical Center, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
46
|
Su H, Rustam YH, Masters CL, Makalic E, McLean CA, Hill AF, Barnham KJ, Reid GE, Vella LJ. Characterization of brain-derived extracellular vesicle lipids in Alzheimer's disease. J Extracell Vesicles 2021; 10:e12089. [PMID: 34012516 PMCID: PMC8111496 DOI: 10.1002/jev2.12089] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
Lipid dyshomeostasis is associated with the most common form of dementia, Alzheimer's disease (AD). Substantial progress has been made in identifying positron emission tomography and cerebrospinal fluid biomarkers for AD, but they have limited use as front-line diagnostic tools. Extracellular vesicles (EVs) are released by all cells and contain a subset of their parental cell composition, including lipids. EVs are released from the brain into the periphery, providing a potential source of tissue and disease specific lipid biomarkers. However, the EV lipidome of the central nervous system is currently unknown and the potential of brain-derived EVs (BDEVs) to inform on lipid dyshomeostasis in AD remains unclear. The aim of this study was to reveal the lipid composition of BDEVs in human frontal cortex, and to determine whether BDEVs have an altered lipid profile in AD. Using semi-quantitative mass spectrometry, we describe the BDEV lipidome, covering four lipid categories, 17 lipid classes and 692 lipid molecules. BDEVs were enriched in glycerophosphoserine (PS) lipids, a characteristic of small EVs. Here we further report that BDEVs are enriched in ether-containing PS lipids, a finding that further establishes ether lipids as a feature of EVs. BDEVs in the AD frontal cortex offered improved detection of dysregulated lipids in AD over global lipid profiling of this brain region. AD BDEVs had significantly altered glycerophospholipid and sphingolipid levels, specifically increased plasmalogen glycerophosphoethanolamine and decreased polyunsaturated fatty acyl containing lipids, and altered amide-linked acyl chain content in sphingomyelin and ceramide lipids relative to CTL. The most prominent alteration was a two-fold decrease in lipid species containing anti-inflammatory/pro-resolving docosahexaenoic acid. The in-depth lipidome analysis provided in this study highlights the advantage of EVs over more complex tissues for improved detection of dysregulated lipids that may serve as potential biomarkers in the periphery.
Collapse
Affiliation(s)
- Huaqi Su
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Department of Biochemistry and PharmacologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Yepy H. Rustam
- Department of Biochemistry and PharmacologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Enes Makalic
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Catriona A. McLean
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Andrew F. Hill
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVictoriaAustralia
| | - Kevin J. Barnham
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Gavin E. Reid
- Department of Biochemistry and PharmacologyThe University of MelbourneParkvilleVictoriaAustralia
- School of Chemistry, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Laura J. Vella
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Department of Surgery, The Royal Melbourne HospitalThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
47
|
Huanglian Jiedu decoction remodels the periphery microenvironment to inhibit Alzheimer's disease progression based on the "brain-gut" axis through multiple integrated omics. ALZHEIMERS RESEARCH & THERAPY 2021; 13:44. [PMID: 33579351 PMCID: PMC7881564 DOI: 10.1186/s13195-021-00779-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND In recent years, excellent results have suggested an association between the "brain-gut" axis and Alzheimer's disease (AD) progression, yet the role of the "brain-gut" axis in AD pathogenesis still remains obscure. Herein, we provided a potential link between the central and peripheral neuroinflammatory disorders in AD progression. METHODS The Morris water maze (MWM) test, immunohistochemistry, ELISA, ProcartaPlex Multiplex immunoassay, multiple LC-MS/MS methods, and the V3-V4 regions of 16S rRNA genes were applied to explore potential biomarkers. RESULTS In Tg-APP/PS1 mice, gut dysbiosis and lipid metabolism were highly associated with AD-like neuroinflammation. The combination of inflammatory factors (IL-6 and INF-γ), phosphatidylcholines (PCs) and SCFA-producing bacteria were expected to be early diagnostic biomarkers for AD. Huanglian Jiedu decoction (HLJDD) suppressed gut dysbiosis and the associated Aβ accumulation, harnessed neuroinflammation and reversed cognitive impairment. CONCLUSION Together, our findings highlighted the roles of neuroinflammation induced by gut dysbiosis and lipid metabolism disorder in AD progression. This integrated metabolomics approach showed its potential to understand the complex mechanisms of HLJDD in the treatment of AD.
Collapse
|
48
|
Jurcau A. The Role of Natural Antioxidants in the Prevention of Dementia-Where Do We Stand and Future Perspectives. Nutrients 2021; 13:282. [PMID: 33498262 PMCID: PMC7909256 DOI: 10.3390/nu13020282] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Dementia, and especially Alzheimer's disease (AD), puts significant burden on global healthcare expenditure through its increasing prevalence. Research has convincingly demonstrated the implication of oxidative stress in the pathogenesis of dementia as well as of the conditions which increase the risk of developing dementia. However, drugs which target single pathways have so far failed in providing significant neuroprotection. Natural antioxidants, due to their effects in multiple pathways through which oxidative stress leads to neurodegeneration and triggers neuroinflammation, could prove valuable weapons in our fight against dementia. Although efficient in vitro and in animal models of AD, natural antioxidants in human trials have many drawbacks related to the limited bioavailability, unknown optimal dose, or proper timing of the treatment. Nonetheless, trials evaluating several of these natural compounds are ongoing, as are attempts to modify these compounds to achieve improved bioavailability.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, nr 1 Universitatii Street, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “Dr. G. Curteanu”, nr 12 Corneliu Coposu Street, 410469 Oradea, Romania
| |
Collapse
|
49
|
Yi Y, Lin Y, Han J, Lee HJ, Park N, Nam G, Park YS, Lee YH, Lim MH. Impact of sphingosine and acetylsphingosines on the aggregation and toxicity of metal-free and metal-treated amyloid-β. Chem Sci 2020; 12:2456-2466. [PMID: 34164011 PMCID: PMC8179336 DOI: 10.1039/d0sc04366d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pathophysiological shifts in the cerebral levels of sphingolipids in Alzheimer's disease (AD) patients suggest a link between sphingolipid metabolism and the disease pathology. Sphingosine (SP), a structural backbone of sphingolipids, is an amphiphilic molecule that is able to undergo aggregation into micelles and micellar aggregates. Considering its structural properties and cellular localization, we hypothesized that SP potentially interacts with amyloid-β (Aβ) and metal ions that are found as pathological components in AD-affected brains, with manifesting its reactivity towards metal-free Aβ and metal-bound Aβ (metal–Aβ). Herein, we report, for the first time, that SP is capable of interacting with both Aβ and metal ions and consequently affects the aggregation of metal-free Aβ and metal–Aβ. Moreover, incubation of SP with Aβ in the absence and presence of metal ions results in the aggravation of toxicity induced by metal-free Aβ and metal–Aβ in living cells. As the simplest acyl derivatives of SP, N-acetylsphingosine and 3-O-acetylsphingosine also influence metal-free Aβ and metal–Aβ aggregation to different degrees, compared to SP. Such slight structural modifications of SP neutralize its ability to exacerbate the cytotoxicity triggered by metal-free Aβ and metal–Aβ. Notably, the reactivity of SP and the acetylsphingosines towards metal-free Aβ and metal–Aβ is determined to be dependent on their formation of micelles and micellar aggregates. Our overall studies demonstrate that SP and its derivatives could directly interact with pathological factors in AD and modify their pathogenic properties at concentrations below and above critical aggregation concentrations. The reactivity of sphingosine and acetylsphingosines towards both metal-free and metal-treated amyloid-β is demonstrated showing a correlation of their micellization properties.![]()
Collapse
Affiliation(s)
- Yelim Yi
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Yuxi Lin
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
| | - Jiyeon Han
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University Gongju 32588 Republic of Korea
| | - Nahye Park
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Geewoo Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Young S Park
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST) Ulsan 44919 Republic of Korea
| | - Young-Ho Lee
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea .,Research Headquarters, Korea Brain Research Institute (KBRI) Daegu 41068 Republic of Korea.,Bio-Analytical Science, University of Science and Technology (UST) Daejeon 34113 Republic of Korea.,Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
50
|
Reshaping circadian metabolism in the suprachiasmatic nucleus and prefrontal cortex by nutritional challenge. Proc Natl Acad Sci U S A 2020; 117:29904-29913. [PMID: 33172990 DOI: 10.1073/pnas.2016589117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Food is a powerful entrainment cue for circadian clocks in peripheral tissues, and changes in the composition of nutrients have been demonstrated to metabolically reprogram peripheral clocks. However, how food challenges may influence circadian metabolism of the master clock in the suprachiasmatic nucleus (SCN) or in other brain areas is poorly understood. Using high-throughput metabolomics, we studied the circadian metabolome profiles of the SCN and medial prefrontal cortex (mPFC) in lean mice compared with mice challenged with a high-fat diet (HFD). Both the mPFC and the SCN displayed a robust cyclic metabolism, with a strikingly high sensitivity to HFD perturbation in an area-specific manner. The phase and amplitude of oscillations were drastically different between the SCN and mPFC, and the metabolic pathways impacted by HFD were remarkably region-dependent. Furthermore, HFD induced a significant increase in the number of cycling metabolites exclusively in the SCN, revealing an unsuspected susceptibility of the master clock to food stress.
Collapse
|