51
|
Yamashita H, Komine O, Fujimori-Tonou N, Yamanaka K. Comprehensive expression analysis with cell-type-specific transcriptome in ALS-linked mutant SOD1 mice: Revisiting the active role of glial cells in disease. Front Cell Neurosci 2022; 16:1045647. [PMID: 36687517 PMCID: PMC9846815 DOI: 10.3389/fncel.2022.1045647] [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: 09/15/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Non-cell autonomous mechanisms are involved in the pathogenesis of amyotrophic lateral sclerosis (ALS), an adult neurodegenerative disease characterized by selective motor neuron loss. While the emerging role of glial cells in ALS has been noted, the detailed cell-type-specific role of glial cells has not been clarified. Here, we examined mRNA expression changes using microarrays of the spinal cords of three distinct lines of mutant superoxide dismutase (SOD) 1 transgenic mice, an established ALS model. Our analysis used a transcriptome database of component cell types in the central nervous system (CNS), as well as SOD1 G93A cell-type transcriptomes. More than half of the differentially expressed genes (DEGs) were highly expressed in microglia, and enrichment analysis of DEGs revealed that immunological reactions were profoundly involved and some transcription factors were upregulated. Our analysis focused on DEGs that are highly expressed in each cell type, as well as chemokines, caspases, and heat shock proteins. Disease-associated microglial genes were upregulated, while homeostatic microglial genes were not, and galectin-3 (Mac2), a known activated microglial marker, was predicted to be ectopically expressed in astrocytes in mutant SOD1 mice. In mutant SOD1 mice, we developed a prediction model for the pathophysiology of different cell types related to TREM2, apolipoprotein E, and lipoproteins. Our analysis offers a viable resource to understand not only the molecular pathologies of each CNS constituent cell type, but also the cellular crosstalk between different cell types under both physiological and pathological conditions in model mice for various neurodegenerative diseases.
Collapse
Affiliation(s)
- Hirofumi Yamashita
- Department of Neurology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan.,Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Noriko Fujimori-Tonou
- Support Unit for Bio-Material Analysis, RRD, RIKEN Center for Brain Science, Wako, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, Japan
| |
Collapse
|
52
|
Aqul AA, Ramirez CM, Lopez AM, Burns DK, Repa JJ, Turley SD. Molecular markers of brain cholesterol homeostasis are unchanged despite a smaller brain mass in a mouse model of cholesteryl ester storage disease. Lipids 2022; 57:3-16. [PMID: 34618372 PMCID: PMC8766890 DOI: 10.1002/lipd.12325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023]
Abstract
Lysosomal acid lipase (LAL), encoded by the gene LIPA, facilitates the intracellular processing of lipids by hydrolyzing cholesteryl esters and triacylglycerols present in newly internalized lipoproteins. Loss-of-function mutations in LIPA result in cholesteryl ester storage disease (CESD) or Wolman disease when mutations cause complete loss of LAL activity. Although the phenotype of a mouse CESD model has been extensively characterized, there has not been a focus on the brain at different stages of disease progression. In the current studies, whole-brain mass and the concentrations of cholesterol in both the esterified (EC) and unesterified (UC) fractions were measured in Lal-/- and matching Lal+/+ mice (FVB-N strain) at ages ranging from 14 up to 280 days after birth. Compared to Lal+/+ controls at 50, 68-76, 140-142, and 230-280 days of age, Lal-/- mice had brain weights that averaged approximately 6%, 7%, 18%, and 20% less, respectively. Brain EC levels were higher in the Lal-/- mice at every age, being elevated 27-fold at 230-280 days. Brain UC concentrations did not show a genotypic difference at any age. The elevated brain EC levels in the Lal-/- mice did not reflect EC in residual blood. An mRNA expression analysis for an array of genes involved in the synthesis, catabolism, storage, and transport of cholesterol in the brains of 141-day old mice did not detect any genotypic differences although the relative mRNA levels for several markers of inflammation were moderately elevated in the Lal-/- mice. The possible sites of EC accretion in the central nervous system are discussed.
Collapse
Affiliation(s)
- Amal A. Aqul
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Charina M. Ramirez
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Adam M. Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Dennis K. Burns
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Joyce J. Repa
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Stephen D. Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| |
Collapse
|
53
|
Frank CJ, McNay EC. Breakdown of the blood-brain barrier: A mediator of increased Alzheimer's risk in patients with metabolic disorders? J Neuroendocrinol 2022; 34:e13074. [PMID: 34904299 PMCID: PMC8791015 DOI: 10.1111/jne.13074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023]
Abstract
Metabolic disorders (MDs), including type 1 and 2 diabetes and chronic obesity, are among the faster growing diseases globally and are a primary risk factor for Alzheimer's disease (AD). The term "type-3 diabetes" has been proposed for AD due to the interrelated cellular, metabolic, and immune features shared by diabetes, insulin resistance (IR), and the cognitive impairment and neurodegeneration found in AD. Patients with MDs and/or AD commonly exhibit altered glucose homeostasis and IR; systemic chronic inflammation encompassing all of the periphery, blood-brain barrier (BBB), and central nervous system; pathological vascular remodeling; and increased BBB permeability that allows transfusion of neurotoxic molecules from the blood to the brain. This review summarizes the components of the BBB, mechanisms through which MDs alter BBB permeability via immune and metabolic pathways, the contribution of BBB dysfunction to the manifestation and progression of AD, and current avenues of therapeutic research that address BBB permeability. In addition, issues with the translational applicability of current animal models of AD regarding BBB dysfunction and proposals for future directions of research that address the relationship between MDs, BBB dysfunction, and AD are discussed.
Collapse
Affiliation(s)
- Corey J Frank
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| | - Ewan C McNay
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| |
Collapse
|
54
|
Structural and Functional Changes of Reconstituted High-Density Lipoprotein (HDL) by Incorporation of α-synuclein: A Potent Antioxidant and Anti-Glycation Activity of α-synuclein and apoA-I in HDL at High Molar Ratio of α-synuclein. Molecules 2021; 26:molecules26247485. [PMID: 34946565 PMCID: PMC8707077 DOI: 10.3390/molecules26247485] [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: 11/10/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 01/05/2023] Open
Abstract
α-synuclein (α-syn) is a major culprit of Parkinson's disease (PD), although lipoprotein metabolism is very important in the pathogenesis of PD. α-syn was expressed and purified using the pET30a expression vector from an E. coli expression system to elucidate the physiological effects of α-syn on lipoprotein metabolism. The human α-syn protein (140 amino acids) with His-tag (8 amino acids) was expressed and purified to at least 95% purity. Isoelectric focusing gel electrophoresis showed that the isoelectric point (pI) of α-syn and apoA-I were pI = 4.5 and pI = 6.4, respectively. The lipid-free α-syn showed almost no phospholipid-binding ability, while apoA-I showed rapid binding ability with a half-time (T1/2) = 8 ± 0.7 min. The α-syn and apoA-I could be incorporated into the reconstituted HDL (rHDL, molar ratio 95:5:1:1, palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC):cholesterol:apoA-I:α-syn with the production of larger particles (92 Å) than apoA-I-rHDL (86 and 78 Å) and α-syn-rHDL (65 Å). An rHDL containing both apoA-I and α-syn showed lower α-helicity around 45% with a red shift of the Trp wavelength maximum fluorescence (WMF) from 339 nm, while apoA-I-HDL showed 76% α-helicity and 337 nm of WMF. The denaturation by urea addition showed that the incorporation of α-syn in rHDL caused a larger increase in the WMF than apoA-I-rHDL, suggesting that the destabilization of the secondary structure of apoA-I by the addition of α-syn. On the other hand, the addition of α-syn induced two-times higher resistance to rHDL glycation at apoA-I:α-syn molar ratios of 1:1 and 1:2. Interestingly, low α-syn in rHDL concentrations, molar ratio of 1:0.5 (apoA-I:α-syn), did not prevent glycation with more multimerization of apoA-I. In the lipid-free and lipid-bound state, α-syn showed more potent antioxidant activity than apoA-I against cupric ion-mediated LDL oxidation. On the other hand, microinjection of α-syn (final 2 μM) resulted in 10% less survival of zebrafish embryos than apoA-I. A subcutaneous injection of α-syn (final 34 μM) resulted in less tail fin regeneration than apoA-I. Interestingly, incorporation of α-syn at a low molar ratio (apoA-I:α-syn, 1:0.5) in rHDL resulted destabilization of the secondary structure and impairment of apoA-I functionality via more oxidation and glycation. However, at a higher molar ratio of α-syn in rHDL (apoA-I:α-syn = 1:1 or 1:2) exhibited potent antioxidant and anti-glycation activity without aggregation. In conclusion, there might be a critical concentration of α-syn and apoA-I in HDL-like complex to prevent the aggregation of apoA-I via structural and functional enhancement.
Collapse
|
55
|
Thiel W, Esposito EJ, Findley AP, Blume ZI, Mitchell DM. Modulation of retinoid-X-receptors differentially regulates expression of apolipoprotein genes apoc1 and apoeb by zebrafish microglia. Biol Open 2021; 11:273656. [PMID: 34878094 PMCID: PMC8822359 DOI: 10.1242/bio.058990] [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: 08/13/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
Transcriptome analyses performed in both human and zebrafish indicate strong expression of Apoe and Apoc1 by microglia. Apoe expression by microglia is well appreciated, but Apoc1 expression has not been well-examined. PPAR/RXR and LXR/RXR receptors appear to regulate expression of the apolipoprotein gene cluster in macrophages, but a similar role in microglia in vivo has not been studied. Here, we characterized microglial expression of apoc1 in the zebrafish central nervous system (CNS) in situ and demonstrate that in the CNS, apoc1 expression is unique to microglia. We then examined the effects of PPAR/RXR and LXR/RXR modulation on microglial expression of apoc1 and apoeb during early CNS development using a pharmacological approach. Changes in apoc1 and apoeb transcripts in response to pharmacological modulation were quantified by RT-qPCR in whole heads, and in individual microglia using hybridization chain reaction (HCR) in situ hybridization. We found that expression of apoc1 and apoeb by microglia were differentially regulated by LXR/RXR and PPAR/RXR modulating compounds, respectively, during development. Our results also suggest RXR receptors could be involved in endogenous induction of apoc1 expression by microglia. Collectively, our work supports the use of zebrafish to better understand regulation and function of these apolipoproteins in the CNS. Summary: Here we investigate expression of two apolipoprotein genes by microglia in the zebrafish model during normal development, and in contexts of pharmacological manipulations that target candidate regulatory receptors.
Collapse
Affiliation(s)
- Whitney Thiel
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Emma J Esposito
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Anna P Findley
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Zachary I Blume
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Diana M Mitchell
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| |
Collapse
|
56
|
Sewell MDE, Jiménez-Sánchez L, Shen X, Edmondson-Stait AJ, Green C, Adams MJ, Rifai OM, McIntosh AM, Lyall DM, Whalley HC, Lawrie SM. Associations between major psychiatric disorder polygenic risk scores and blood-based markers in UK biobank. Brain Behav Immun 2021; 97:32-41. [PMID: 34107350 DOI: 10.1016/j.bbi.2021.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/16/2021] [Accepted: 06/04/2021] [Indexed: 01/08/2023] Open
Abstract
Major depressive disorder (MDD), schizophrenia (SCZ), and bipolar disorder (BD) have both shared and discrete genetic risk factors, and are associated with peripheral abnormalities. The relationships between such genetic architectures and blood-based markers are, however, unclear. We investigated relationships between polygenic risk scores (PRS) for these disorders and peripheral markers in the UK Biobank cohort. We calculated polygenic risk scores for n = 367,329 (MDD PRS), n = 366,465 (SCZ PRS), and n = 366,383 (BD PRS) UK Biobank cohort subjects. We then examined associations between disorder PRS and 58 inflammatory/immune, hematological, bone, cardiovascular, hormone, liver, renal and diabetes-associated blood markers using two generalized linear regression models: 'minimally adjusted' controlling for variables such as age and sex, and 'fully adjusted' including additional lifestyle covariates: BMI, alcohol and smoking status, and medication intake. There were 38/58 MDD PRS, 32/58 SCZ PRS, and 20/58 BD PRS-blood marker associations detected for our minimally adjusted model. Of these, 13/38 (MDD PRS), 14/32 (SCZ PRS), and 10/20 (BD PRS) associations remained significant after controlling for lifestyle factors. Many were disorder-specific, with 8/13 unique MDD PRS associations identified. Several disorder-specific associations for MDD and SCZ were immune-related, with mostly positive and negative associations identified for MDD and SCZ PRS respectively. This study suggests that MDD, SCZ and BD have both shared and distinct peripheral markers associated with disorder-specific genetic risk. The results also implicate inflammatory dysfunction in MDD and SCZ, albeit with differences in patterns between the two conditions, and enrich our understanding of potential underlying pathophysiological mechanisms in major psychiatric disorders.
Collapse
Affiliation(s)
- Michael D E Sewell
- Translational Neuroscience PhD Programme, Centre for Clinical Brain Sciences, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK.
| | - Lorena Jiménez-Sánchez
- Translational Neuroscience PhD Programme, Centre for Clinical Brain Sciences, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - Xueyi Shen
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - Amelia J Edmondson-Stait
- Translational Neuroscience PhD Programme, Centre for Clinical Brain Sciences, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - Claire Green
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Olivia M Rifai
- Translational Neuroscience PhD Programme, Centre for Clinical Brain Sciences, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Donald M Lyall
- Institute of Health & Wellbeing, University of Glasgow, Glasgow G12 8RZ, UK
| | - Heather C Whalley
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| | - Stephen M Lawrie
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh EH10 5HF, UK
| |
Collapse
|
57
|
Picard C, Nilsson N, Labonté A, Auld D, Rosa-Neto P, Ashton NJ, Zetterberg H, Blennow K, Breitner JCB, Villeneuve S, Poirier J. Apolipoprotein B is a novel marker for early tau pathology in Alzheimer's disease. Alzheimers Dement 2021; 18:875-887. [PMID: 34590423 PMCID: PMC9293308 DOI: 10.1002/alz.12442] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 01/08/2023]
Abstract
INTRODUCTION We examine the role of brain apolipoprotein B (apoB) as a putative marker of early tau pathology and cognitive decline. METHODS Cerebrospinal fluid (CSF) samples from cognitively normal and Alzheimer's disease (AD) participants were collected to measure protein levels of apoB and AD biomarkers amyloid beta (Aβ), t-tau and p-tau, as well as synaptic markers GAP43, SYNAPTOTAGMIN-1, synaptosome associated protein 25 (SNAP-25), and NEUROGRANIN. CSF apoB levels were contrasted with positron emission tomography (PET) scan measures of Aβ (18F-NAV4694) and Tau (flortaucipir) along with cognitive assessment alterations over 6 to 8 years. RESULTS CSF apoB levels were elevated in AD participants and correlated with t-tau, p-tau, and the four synaptic markers in pre-symptomatic individuals. In the latter, CSF apoB levels correlated with PET flortaucipir-binding in entorhinal, parahippocampal, and fusiform regions. Baseline CSF apoB levels were associated with longitudinal visuospatial cognitive decline. DISCUSSION CSF apoB markedly associates with early tau dysregulation in asymptomatic subjects and identifies at-risk individuals predisposed to develop visuospatial cognitive decline over time.
Collapse
Affiliation(s)
- Cynthia Picard
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Nathalie Nilsson
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | | | - Pedro Rosa-Neto
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | -
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - John C B Breitner
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | -
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| |
Collapse
|
58
|
Van Valkenburgh J, Meuret C, Martinez AE, Kodancha V, Solomon V, Chen K, Yassine HN. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front Physiol 2021; 12:700847. [PMID: 34552500 PMCID: PMC8450374 DOI: 10.3389/fphys.2021.700847] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
High-density lipoproteins (HDLs) are complex, heterogenous lipoprotein particles, consisting of a large family of apolipoproteins, formed in subspecies of distinct shapes, sizes, and functions and are synthesized in both the brain and the periphery. HDL apolipoproteins are important determinants of Alzheimer’s disease (AD) pathology and vascular dementia, having both central and peripheral effects on brain amyloid-beta (Aβ) accumulation and vascular functions, however, the extent to which HDL particles (HLD-P) can exchange their protein and lipid components between the central nervous system (CNS) and the systemic circulation remains unclear. In this review, we delineate how HDL’s structure and composition enable exchange between the brain, cerebrospinal fluid (CSF) compartment, and vascular cells that ultimately affect brain amyloid metabolism and atherosclerosis. Accordingly, we then elucidate how modifications of HDL-P have diagnostic and therapeutic potential for brain vascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Cristiana Meuret
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ashley E Martinez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vibha Kodancha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
59
|
Interactions of Lipids, Lipoproteins, and Apolipoproteins with the Blood-Brain Barrier. Pharm Res 2021; 38:1469-1475. [PMID: 34518942 DOI: 10.1007/s11095-021-03098-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/22/2021] [Indexed: 10/20/2022]
Abstract
Lipids and lipoproteins are a diverse group of substances and their interactions with the blood-brain barrier (BBB) is similarly diverse. Some lipoproteins such as high density lipoprotein (HDL), apolipoprotein (apo) A-I, apoJ, some free fatty acids, and triglycerides cross the BBB whereas others such as apoE do not. Some forms of cholesterol can cross the BBB and others do not. Lipids can have effects on BBB preservation and function: HDL may protect the BBB during multiple sclerosis, cholesterol can disrupt the BBB, and triglycerides inhibit the transport of leptin across the BBB and the activation of the hypothalamic leptin receptor. ApoE is associated with many effects on the BBB, with the specific isoform apoE4 having detrimental effects. In summary, the diverse ways in which lipids, lipoproteins, and apolipoproteins interact with the BBB is important in both health and disease.
Collapse
|
60
|
Berland C, Small DM, Luquet S, Gangarossa G. Dietary lipids as regulators of reward processes: multimodal integration matters. Trends Endocrinol Metab 2021; 32:693-705. [PMID: 34148784 DOI: 10.1016/j.tem.2021.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/08/2021] [Accepted: 05/24/2021] [Indexed: 02/03/2023]
Abstract
The abundance of energy-dense and palatable diets in the modern food environment tightly contributes to the obesity pandemic. The reward circuit participates to the regulation of body homeostasis by integrating energy-related signals with neural substrates encoding cognitive and motivational components of feeding behaviors. Obesity and lipid-rich diets alter dopamine (DA) transmission leading to reward dysfunctions and food overconsumption. Recent reports indicate that dietary lipids can act, directly and indirectly, as functional modulators of the DA circuit. This raises the possibility that nutritional or genetic conditions affecting 'lipid sensing' mechanisms might lead to maladaptations of the DA system. Here, we discuss the most recent findings connecting dietary lipid sensing with DA signaling and its multimodal influence on circuits regulating food-reward processes.
Collapse
Affiliation(s)
- Chloé Berland
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; Department of Medicine, The Naomi Berrie Diabetes Center, Columbia University, New York, NY 10032, USA
| | - Dana M Small
- Department of Psychiatry, and the Modern Diet and Physiology Research Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Serge Luquet
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France.
| | | |
Collapse
|
61
|
Chai AB, Lam HHJ, Kockx M, Gelissen IC. Apolipoprotein E isoform-dependent effects on the processing of Alzheimer's amyloid-β. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158980. [PMID: 34044125 DOI: 10.1016/j.bbalip.2021.158980] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022]
Abstract
Since the identification of the apolipoprotein E (apoE) *ε4 allele as a major genetic risk factor for late-onset Alzheimer's disease, significant efforts have been aimed at elucidating how apoE4 expression confers greater brain amyloid-β (Aβ) burden, earlier disease onset and worse clinical outcomes compared to apoE2 and apoE3. ApoE primarily functions as a lipid carrier to regulate cholesterol metabolism in circulation as well as in the brain. However, it has also been suggested to interact with hydrophobic Aβ peptides to influence their processing in an isoform-dependent manner. Here, we review evidence from in vitro and in vivo studies extricating the effects of the three apoE isoforms, on different stages of the Aβ processing pathway including synthesis, aggregation, deposition, clearance and degradation. ApoE4 consistently correlates with impaired Aβ clearance, however data regarding Aβ synthesis and aggregation are conflicting and likely reflect inconsistencies in experimental approaches across studies. We further discuss the physical and chemical properties of apoE that may explain the inherent differences in activity between the isoforms. The lipidation status and lipid transport function of apoE are intrinsically linked with its ability to interact with Aβ. Traditionally, apoE-oriented therapeutic strategies for Alzheimer's disease have been proposed to non-specifically enhance or inhibit apoE activity. However, given the wide-ranging physiological functions of apoE in the brain and periphery, a more viable approach may be to specifically target and neutralise the pathological apoE4 isoform.
Collapse
Affiliation(s)
- Amanda B Chai
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Hin Hei Julian Lam
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Maaike Kockx
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Concord, NSW 2139, Australia
| | - Ingrid C Gelissen
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
62
|
Lam M, Chen CY, Ge T, Xia Y, Hill DW, Trampush JW, Yu J, Knowles E, Davies G, Stahl EA, Huckins L, Liewald DC, Djurovic S, Melle I, Christoforou A, Reinvang I, DeRosse P, Lundervold AJ, Steen VM, Espeseth T, Räikkönen K, Widen E, Palotie A, Eriksson JG, Giegling I, Konte B, Hartmann AM, Roussos P, Giakoumaki S, Burdick KE, Payton A, Ollier W, Chiba-Falek O, Koltai DC, Need AC, Cirulli ET, Voineskos AN, Stefanis NC, Avramopoulos D, Hatzimanolis A, Smyrnis N, Bilder RM, Freimer NB, Cannon TD, London E, Poldrack RA, Sabb FW, Congdon E, Conley ED, Scult MA, Dickinson D, Straub RE, Donohoe G, Morris D, Corvin A, Gill M, Hariri AR, Weinberger DR, Pendleton N, Bitsios P, Rujescu D, Lahti J, Le Hellard S, Keller MC, Andreassen OA, Deary IJ, Glahn DC, Huang H, Liu C, Malhotra AK, Lencz T. Identifying nootropic drug targets via large-scale cognitive GWAS and transcriptomics. Neuropsychopharmacology 2021; 46:1788-1801. [PMID: 34035472 PMCID: PMC8357785 DOI: 10.1038/s41386-021-01023-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/22/2021] [Accepted: 04/12/2021] [Indexed: 02/05/2023]
Abstract
Broad-based cognitive deficits are an enduring and disabling symptom for many patients with severe mental illness, and these impairments are inadequately addressed by current medications. While novel drug targets for schizophrenia and depression have emerged from recent large-scale genome-wide association studies (GWAS) of these psychiatric disorders, GWAS of general cognitive ability can suggest potential targets for nootropic drug repurposing. Here, we (1) meta-analyze results from two recent cognitive GWAS to further enhance power for locus discovery; (2) employ several complementary transcriptomic methods to identify genes in these loci that are credibly associated with cognition; and (3) further annotate the resulting genes using multiple chemoinformatic databases to identify "druggable" targets. Using our meta-analytic data set (N = 373,617), we identified 241 independent cognition-associated loci (29 novel), and 76 genes were identified by 2 or more methods of gene identification. Actin and chromatin binding gene sets were identified as novel pathways that could be targeted via drug repurposing. Leveraging our transcriptomic and chemoinformatic databases, we identified 16 putative genes targeted by existing drugs potentially available for cognitive repurposing.
Collapse
Affiliation(s)
- Max Lam
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Institute of Mental Health, Singapore, Singapore
| | - Chia-Yen Chen
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Biogen, Inc, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Tian Ge
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Yan Xia
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Psychiatry Department, SUNY Upstate Medical University, Syracuse, NY, USA
| | - David W Hill
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland, UK
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Joey W Trampush
- Department of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jin Yu
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
| | - Emma Knowles
- Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Olin Neuropsychic Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - Gail Davies
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland, UK
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Eli A Stahl
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Huckins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David C Liewald
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ingrid Melle
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Andrea Christoforou
- Spaulding Rehabilitation Hospital Boston, Charlestown, MA, USA
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ivar Reinvang
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Pamela DeRosse
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Astri J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Vidar M Steen
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Thomas Espeseth
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Johan G Eriksson
- Department of General Practice, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhälsan Research Center, Helsinki, Finland
| | - Ina Giegling
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Bettina Konte
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Annette M Hartmann
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2), James J. Peters VA Medical Center, Bronx, NY, USA
| | | | - Katherine E Burdick
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education, and Clinical Center (VISN 2), James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Psychiatry - Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Antony Payton
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, The University of Manchester, Manchester, UK
| | - William Ollier
- Centre for Epidemiology, Division of Population Health, Health Services Research & Primary Care, The University of Manchester, Manchester, UK
- School of Healthcare Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Bryan Alzheimer's Disease Research Center, and Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Deborah C Koltai
- Psychiatry and Behavioral Sciences, Division of Medical Psychology, and Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Anna C Need
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Aristotle N Voineskos
- Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Nikos C Stefanis
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens Medical School, University General Hospital "ATTIKON", Athens, Greece
- University Mental Health Research Institute, Athens, Greece
- Neurobiology Research Institute, Theodor-Theohari Cozzika Foundation, Athens, Greece
| | - Dimitrios Avramopoulos
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex Hatzimanolis
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens Medical School, University General Hospital "ATTIKON", Athens, Greece
- University Mental Health Research Institute, Athens, Greece
- Neurobiology Research Institute, Theodor-Theohari Cozzika Foundation, Athens, Greece
| | - Nikolaos Smyrnis
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens Medical School, University General Hospital "ATTIKON", Athens, Greece
- University Mental Health Research Institute, Athens, Greece
| | - Robert M Bilder
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - Nelson B Freimer
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - Tyrone D Cannon
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Edythe London
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | | | - Fred W Sabb
- Robert and Beverly Lewis Center for Neuroimaging, University of Oregon, Eugene, OR, USA
| | - Eliza Congdon
- UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, USA
| | | | - Matthew A Scult
- Weill Cornell Psychiatry at NewYork-Presbyterian, Weill Cornell Medical Center, New York, NY, USA
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
| | - Dwight Dickinson
- Clinical and Translational Neuroscience Branch, Intramural Research Program, National Institute of Mental Health, National Institute of Health, Bethesda, MD, USA
| | - Richard E Straub
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Gary Donohoe
- Neuroimaging, Cognition & Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland, Galway, Ireland
| | - Derek Morris
- Neuroimaging, Cognition & Genomics (NICOG) Centre, School of Psychology and Discipline of Biochemistry, National University of Ireland, Galway, Ireland
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Michael Gill
- Neuropsychiatric Genetics Research Group, Department of Psychiatry and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Ahmad R Hariri
- Laboratory of NeuroGenetics, Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Neil Pendleton
- Division of Neuroscience and Experimental Psychology/School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Panos Bitsios
- Department of Psychiatry and Behavioral Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, GR, Greece
| | - Dan Rujescu
- Department of Psychiatry, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
| | - Stephanie Le Hellard
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Matthew C Keller
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ian J Deary
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, Scotland, UK
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - David C Glahn
- Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Olin Neuropsychic Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| | - Hailiang Huang
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Psychiatry Department, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Anil K Malhotra
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Todd Lencz
- Division of Psychiatry Research, The Zucker Hillside Hospital, Glen Oaks, NY, USA.
- Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA.
- Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
63
|
Lee SE, Yang H, Sung Y, Kim Y, Park SA. Region-Specific Differences in the Apoe4-dependent Response to Focal Brain Injury. Exp Neurobiol 2021; 30:285-293. [PMID: 34376629 PMCID: PMC8424383 DOI: 10.5607/en21022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/16/2021] [Accepted: 08/04/2021] [Indexed: 11/19/2022] Open
Abstract
Apolipoprotein E (apoE) plays a role in various physiological functions including lipid transport, synaptic plasticity, and immune modulation. Epidemiological studies suggest that the apoE4 allele increases the risk of post-traumatic sequelae. This study was performed to investigate regionspecific effects of the apoE4 isoform on post-traumatic neurodegeneration. Two focal brain injuries were introduced separately in the motor cortex and hippocampus of apoE4 knock-in, apoE3 knock-in, apoE knockout, and wild-type (WT) mice. Western blotting showed that the expression levels of pre-synaptic and post-synaptic markers at the recovery stage were lower in the hippocampal injury core in apoE4 mice, compared with apoE3 and WT mice. Fast glial activation (determined by immunohistochemistry with glial fibrillary acidic protein, ionized calcium binding adaptor molecule 1, and cluster of differentiation 45 antibodies) was characteristic of apoE4 mice with hippocampal injury penumbra. apoE4-specific changes were not observed after cortical injury. The intensity of microglial activation in the hippocampus was inversely correlated with the volume of injury reduction on sequential magnetic resonance imaging examinations, when validated using matched samples. These findings indicate that the effects of the interaction between apoE4 and focal brain damage are specific to the hippocampus. Manipulation of inflammatory cell responses could be beneficial for reducing post-traumatic hippocampal neurodegeneration in apoE4 carriers.
Collapse
Affiliation(s)
- Sung Eun Lee
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine
| | - Haijie Yang
- Department of Pharmacology, Ajou University School of Medicine
| | | | - Younghoon Kim
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine
| | - Sun Ah Park
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine
- Department of Neurology, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
64
|
Demetci P, Cheng W, Darnell G, Zhou X, Ramachandran S, Crawford L. Multi-scale inference of genetic trait architecture using biologically annotated neural networks. PLoS Genet 2021; 17:e1009754. [PMID: 34411094 PMCID: PMC8407593 DOI: 10.1371/journal.pgen.1009754] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 08/31/2021] [Accepted: 07/31/2021] [Indexed: 01/01/2023] Open
Abstract
In this article, we present Biologically Annotated Neural Networks (BANNs), a nonlinear probabilistic framework for association mapping in genome-wide association (GWA) studies. BANNs are feedforward models with partially connected architectures that are based on biological annotations. This setup yields a fully interpretable neural network where the input layer encodes SNP-level effects, and the hidden layer models the aggregated effects among SNP-sets. We treat the weights and connections of the network as random variables with prior distributions that reflect how genetic effects manifest at different genomic scales. The BANNs software uses variational inference to provide posterior summaries which allow researchers to simultaneously perform (i) mapping with SNPs and (ii) enrichment analyses with SNP-sets on complex traits. Through simulations, we show that our method improves upon state-of-the-art association mapping and enrichment approaches across a wide range of genetic architectures. We then further illustrate the benefits of BANNs by analyzing real GWA data assayed in approximately 2,000 heterogenous stock of mice from the Wellcome Trust Centre for Human Genetics and approximately 7,000 individuals from the Framingham Heart Study. Lastly, using a random subset of individuals of European ancestry from the UK Biobank, we show that BANNs is able to replicate known associations in high and low-density lipoprotein cholesterol content.
Collapse
Affiliation(s)
- Pinar Demetci
- Department of Computer Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Wei Cheng
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
| | - Gregory Darnell
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sohini Ramachandran
- Department of Computer Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
| | - Lorin Crawford
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Microsoft Research New England, Cambridge, Massachusetts, United States of America
- Department of Biostatistics, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
65
|
Characterization of the Striatal Extracellular Matrix in a Mouse Model of Parkinson's Disease. Antioxidants (Basel) 2021; 10:antiox10071095. [PMID: 34356328 PMCID: PMC8301085 DOI: 10.3390/antiox10071095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 02/02/2023] Open
Abstract
Parkinson’s disease’s etiology is unknown, although evidence suggests the involvement of oxidative modifications of intracellular components in disease pathobiology. Despite the known involvement of the extracellular matrix in physiology and disease, the influence of oxidative stress on the matrix has been neglected. The chemical modifications that might accumulate in matrix components due to their long half-live and the low amount of extracellular antioxidants could also contribute to the disease and explain ineffective cellular therapies. The enriched striatal extracellular matrix from a mouse model of Parkinson’s disease was characterized by Raman spectroscopy. We found a matrix fingerprint of increased oxalate content and oxidative modifications. To uncover the effects of these changes on brain cells, we morphologically characterized the primary microglia used to repopulate this matrix and further quantified the effects on cellular mechanical stress by an intracellular fluorescence resonance energy transfer (FRET)-mechanosensor using the U-2 OS cell line. Our data suggest changes in microglia survival and morphology, and a decrease in cytoskeletal tension in response to the modified matrix from both hemispheres of 6-hydroxydopamine (6-OHDA)-lesioned animals. Collectively, these data suggest that the extracellular matrix is modified, and underscore the need for its thorough investigation, which may reveal new ways to improve therapies or may even reveal new therapies.
Collapse
|
66
|
Ralhan I, Chang CL, Lippincott-Schwartz J, Ioannou MS. Lipid droplets in the nervous system. J Cell Biol 2021; 220:e202102136. [PMID: 34152362 PMCID: PMC8222944 DOI: 10.1083/jcb.202102136] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 01/20/2023] Open
Abstract
Lipid droplets are dynamic intracellular lipid storage organelles that respond to the physiological state of cells. In addition to controlling cell metabolism, they play a protective role for many cellular stressors, including oxidative stress. Despite prior descriptions of lipid droplets appearing in the brain as early as a century ago, only recently has the role of lipid droplets in cells found in the brain begun to be understood. Lipid droplet functions have now been described for cells of the nervous system in the context of development, aging, and an increasing number of neuropathologies. Here, we review the basic mechanisms of lipid droplet formation, turnover, and function and discuss how these mechanisms enable lipid droplets to function in different cell types of the nervous system under healthy and pathological conditions.
Collapse
Affiliation(s)
- Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Chi-Lun Chang
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | | | - Maria S. Ioannou
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
67
|
Phillips W, Willms E, Hill AF. Understanding extracellular vesicle and nanoparticle heterogeneity: Novel methods and considerations. Proteomics 2021; 21:e2000118. [PMID: 33857352 PMCID: PMC8365743 DOI: 10.1002/pmic.202000118] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of membrane-enclosed nanoparticles released by cells. They play a role in intercellular communication and are involved in numerous physiological and pathological processes. Cells release subpopulations of EVs with distinct composition and inherent biological function which overlap in size. Current size-based isolation methods are, therefore, not optimal to discriminate between functional EV subpopulations. In addition, EVs overlap in size with several other biological nanoparticles, such as lipoproteins and viruses. Proteomic analysis has allowed for more detailed study of EV composition, and EV isolation approaches based on this could provide a promising alternative for purification based on size. Elucidating EV heterogeneity and the characteristics and role of EV subpopulations will advance our understanding of EV biology and the role of EVs in health and disease. Here, we discuss current knowledge of EV composition, EV heterogeneity and advances in affinity based EV isolation tools.
Collapse
Affiliation(s)
- William Phillips
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Eduard Willms
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Andrew F. Hill
- Department of Biochemistry and GeneticsLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| |
Collapse
|
68
|
Li N, Zhou P, Tang H, He L, Fang X, Zhao J, Wang X, Qi Y, Sun C, Lin Y, Qin F, Yang M, Zhang Z, Liao C, Zheng S, Peng X, Xue T, Zhu Q, Li H, Li Y, Liu L, Huang J, Liu L, Peng C, Kaindl AM, Gecz J, Han D, Liu D, Xu K, Hu H. In-depth analysis reveals complex molecular aetiology in a cohort of idiopathic cerebral palsy. Brain 2021; 145:119-141. [PMID: 34077496 PMCID: PMC8967106 DOI: 10.1093/brain/awab209] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 12/02/2022] Open
Abstract
Cerebral palsy is the most prevalent physical disability in children; however, its inherent molecular mechanisms remain unclear. In the present study, we performed in-depth clinical and molecular analysis on 120 idiopathic cerebral palsy families, and identified underlying detrimental genetic variants in 45% of these patients. In addition to germline variants, we found disease-related postzygotic mutations in ∼6.7% of cerebral palsy patients. We found that patients with more severe motor impairments or a comorbidity of intellectual disability had a significantly higher chance of harbouring disease-related variants. By a compilation of 114 known cerebral-palsy-related genes, we identified characteristic features in terms of inheritance and function, from which we proposed a dichotomous classification system according to the expression patterns of these genes and associated cognitive impairments. In two patients with both cerebral palsy and intellectual disability, we revealed that the defective TYW1, a tRNA hypermodification enzyme, caused primary microcephaly and problems in motion and cognition by hindering neuronal proliferation and migration. Furthermore, we developed an algorithm and demonstrated in mouse brains that this malfunctioning hypermodification specifically perturbed the translation of a subset of proteins involved in cell cycling. This finding provided a novel and interesting mechanism for congenital microcephaly. In another cerebral palsy patient with normal intelligence, we identified a mitochondrial enzyme GPAM, the hypomorphic form of which led to hypomyelination of the corticospinal tract in both human and mouse models. In addition, we confirmed that the aberrant Gpam in mice perturbed the lipid metabolism in astrocytes, resulting in suppressed astrocytic proliferation and a shortage of lipid contents supplied for oligodendrocytic myelination. Taken together, our findings elucidate novel aspects of the aetiology of cerebral palsy and provide insights for future therapeutic strategies.
Collapse
Affiliation(s)
- Na Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Pei Zhou
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120, Guangzhou, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120, Guangzhou, China
| | - Xiang Fang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Jinxiang Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001, Nantong, China
| | - Xin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001, Nantong, China
| | - Yifei Qi
- Division of Uterine Vascular Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Chuanbo Sun
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Yunting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Fengying Qin
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Miaomiao Yang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Zhan Zhang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Caihua Liao
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Shuxin Zheng
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Xiaofang Peng
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Ting Xue
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Qianying Zhu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Hong Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Yan Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120, Guangzhou, China
| | - Jingyu Huang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120, Guangzhou, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Changgeng Peng
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, 200029, Shanghai, China
| | - Angela M Kaindl
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin, 13353, Berlin, Germany.,Department of Pediatric Neurology, Charité-Universitätsmedizin, 13353, Berlin, Germany.,Center for Chronically Sick Children, Charité-Universitätsmedizin, 13353, Berlin, Germany
| | - Jozef Gecz
- Adelaide Medical School, University of Adelaide, SA5005, Adelaide, Australia
| | - Dingding Han
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Dong Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, 226001, Nantong, China
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510120, Guangzhou, China
| | - Hao Hu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China.,Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China.,Third Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| |
Collapse
|
69
|
Azubuike-Osu SO, Famurewa AC, David JC, Abi I, Ogbu PN, Oparaji CK, Nwaeze KG, Akunna GG. Virgin Coconut Oil Resists Arsenic-Induced Cerebral Neurotoxicity and Cholesterol Imbalance via Suppression of Oxidative Stress, Adenosine Deaminase and Acetylcholinesterase Activities in Rats. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211016962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Arsenic (As) is a classic neurotoxicant; its pathogenesis is associated with oxidative stress and oxidative stress-mediated cholinergic deficits. This study explored antioxidant activity of virgin coconut oil (VCO) against sodium arsenite-induced oxidative stress-mediated cerebral neurotoxicity in rats. Eighteen rats were divided into 3 groups- Normal control, As control and VCO + As. The VCO (5 mL/kg) was given once daily by oral gavage from day 1 to day 21, while As (10 mg/kg) was given once daily by oral gavage from day 15 to day 21. Cerebral superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), malondialdehyde (MDA), adenosine deaminase (ADA) and acetylcholinesterase (AchE) activities were analysed. Nitric oxide (NO), lipid profile, phospholipid (PL), and reduced glutathione (GSH) were also evaluated in cerebral homogenate. The cerebrum was sectioned for histological analysis. Administration of As induced significant depressions in antioxidant enzymes, GSH, PL, and HDL-c compared to normal control. Levels of MDA, NO, total cholesterol and activities of ADA, AchE in the cerebrum were markedly increased by As compared to normal rats. Lipid profile indices and PL were prominently altered by As. Histopathological study supported the biochemical findings through extensive cerebral damage. In contrast, oral supplementation of VCO prior to and along with As treatment significantly attenuated the As-induced biochemical alterations and restored near-normal histology. VCO attenuates cerebral neurotoxicity by strengthening endogenous antioxidant defence and cholinergic function via counteracting free-radical-mediated arsenic toxicity.
Collapse
Affiliation(s)
- Sharon O. Azubuike-Osu
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Ademola C. Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Japheth C. David
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Innocent Abi
- Department of Physiology, Benue State University, Makurdi, Nigeria
| | - Patience N. Ogbu
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex-Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Chiedozie K. Oparaji
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Konyefom G. Nwaeze
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
| | - Godson G. Akunna
- Department of Anatomy, College of Medicine and Health Sciences, Bowen University, Nigeria
| |
Collapse
|
70
|
Folick A, Koliwad SK, Valdearcos M. Microglial Lipid Biology in the Hypothalamic Regulation of Metabolic Homeostasis. Front Endocrinol (Lausanne) 2021; 12:668396. [PMID: 34122343 PMCID: PMC8191416 DOI: 10.3389/fendo.2021.668396] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/05/2021] [Indexed: 12/18/2022] Open
Abstract
In mammals, myeloid cells help maintain the homeostasis of peripheral metabolic tissues, and their immunologic dysregulation contributes to the progression of obesity and associated metabolic disease. There is accumulating evidence that innate immune cells also serve as functional regulators within the mediobasal hypothalamus (MBH), a critical brain region controlling both energy and glucose homeostasis. Specifically, microglia, the resident parenchymal myeloid cells of the CNS, play important roles in brain physiology and pathology. Recent studies have revealed an expanding array of microglial functions beyond their established roles as immune sentinels, including roles in brain development, circuit refinement, and synaptic organization. We showed that microglia modulate MBH function by transmitting information resulting from excess nutrient consumption. For instance, microglia can sense the excessive consumption of saturated fats and instruct neurons within the MBH accordingly, leading to responsive alterations in energy balance. Interestingly, the recent emergence of high-resolution single-cell techniques has enabled specific microglial populations and phenotypes to be profiled in unprecedented detail. Such techniques have highlighted specific subsets of microglia notable for their capacity to regulate the expression of lipid metabolic genes, including lipoprotein lipase (LPL), apolipoprotein E (APOE) and Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). The discovery of this transcriptional signature highlights microglial lipid metabolism as a determinant of brain health and disease pathogenesis, with intriguing implications for the treatment of brain disorders and potentially metabolic disease. Here we review our current understanding of how changes in microglial lipid metabolism could influence the hypothalamic control of systemic metabolism.
Collapse
Affiliation(s)
- Andrew Folick
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Suneil K. Koliwad
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
71
|
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
|
72
|
Yin J, Spillman E, Cheng ES, Short J, Chen Y, Lei J, Gibbs M, Rosenthal JS, Sheng C, Chen YX, Veerasammy K, Choetso T, Abzalimov R, Wang B, Han C, He Y, Yuan Q. Brain-specific lipoprotein receptors interact with astrocyte derived apolipoprotein and mediate neuron-glia lipid shuttling. Nat Commun 2021; 12:2408. [PMID: 33893307 PMCID: PMC8065144 DOI: 10.1038/s41467-021-22751-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/29/2021] [Indexed: 02/08/2023] Open
Abstract
Lipid shuttling between neurons and glia contributes to the development, function, and stress responses of the nervous system. To understand how a neuron acquires its lipid supply from specific lipoproteins and their receptors, we perform combined genetic, transcriptome, and biochemical analyses in the developing Drosophila larval brain. Here we report, the astrocyte-derived secreted lipocalin Glial Lazarillo (GLaz), a homolog of human Apolipoprotein D (APOD), and its neuronal receptor, the brain-specific short isoforms of Drosophila lipophorin receptor 1 (LpR1-short), cooperatively mediate neuron-glia lipid shuttling and support dendrite morphogenesis. The isoform specificity of LpR1 defines its distribution, binding partners, and ability to support proper dendrite growth and synaptic connectivity. By demonstrating physical and functional interactions between GLaz/APOD and LpR1, we elucidate molecular pathways mediating lipid trafficking in the fly brain, and provide in vivo evidence indicating isoform-specific expression of lipoprotein receptors as a key mechanism for regulating cell-type specific lipid recruitment.
Collapse
Affiliation(s)
- Jun Yin
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Emma Spillman
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Ethan S Cheng
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jacob Short
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Yang Chen
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jingce Lei
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mary Gibbs
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Justin S Rosenthal
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Chengyu Sheng
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuki X Chen
- The City University of New York, Graduate Center-Advanced Science Research Center, New York, NY, USA
- The City College of New York, CUNY, New York, NY, USA
| | - Kelly Veerasammy
- The City University of New York, Graduate Center-Advanced Science Research Center, New York, NY, USA
- The City College of New York, CUNY, New York, NY, USA
| | - Tenzin Choetso
- The City University of New York, Graduate Center-Advanced Science Research Center, New York, NY, USA
- The City College of New York, CUNY, New York, NY, USA
| | - Rinat Abzalimov
- The City University of New York, Graduate Center-Advanced Science Research Center, New York, NY, USA
| | - Bei Wang
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Chun Han
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Ye He
- The City University of New York, Graduate Center-Advanced Science Research Center, New York, NY, USA
| | - Quan Yuan
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
73
|
Mutlu AS, Duffy J, Wang MC. Lipid metabolism and lipid signals in aging and longevity. Dev Cell 2021; 56:1394-1407. [PMID: 33891896 DOI: 10.1016/j.devcel.2021.03.034] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/05/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Lipids play crucial roles in regulating aging and longevity. In the past few decades, a series of genetic pathways have been discovered to regulate lifespan in model organisms. Interestingly, many of these regulatory pathways are linked to lipid metabolism and lipid signaling. Lipid metabolic enzymes undergo significant changes during aging and are regulated by different longevity pathways. Lipids also actively modulate lifespan and health span as signaling molecules. In this review, we summarize recent insights into the roles of lipid metabolism and lipid signaling in aging and discuss lipid-related interventions in promoting longevity.
Collapse
Affiliation(s)
- Ayse Sena Mutlu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jonathon Duffy
- Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
74
|
Mendonça MCP, Kont A, Aburto MR, Cryan JF, O'Driscoll CM. Advances in the Design of (Nano)Formulations for Delivery of Antisense Oligonucleotides and Small Interfering RNA: Focus on the Central Nervous System. Mol Pharm 2021; 18:1491-1506. [PMID: 33734715 PMCID: PMC8824433 DOI: 10.1021/acs.molpharmaceut.0c01238] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
RNA-based therapeutics have emerged
as one of the most powerful
therapeutic options used for the modulation of gene/protein expression
and gene editing with the potential to treat neurodegenerative diseases.
However, the delivery of nucleic acids to the central nervous system
(CNS), in particular by the systemic route, remains a major hurdle.
This review will focus on the strategies for systemic delivery of
therapeutic nucleic acids designed to overcome these barriers. Pathways
and mechanisms of transport across the blood–brain barrier
which could be exploited for delivery are described, focusing in particular
on smaller nucleic acids including antisense oligonucleotides (ASOs)
and small interfering RNA (siRNA). Approaches used to enhance delivery
including chemical modifications, nanocarrier systems, and target
selection (cell-specific delivery) are critically analyzed. Learnings
achieved from a comparison of the successes and failures reported
for CNS delivery of ASOs versus siRNA will help identify opportunities
for a wider range of nucleic acids and accelerate the clinical translation
of these innovative therapies.
Collapse
Affiliation(s)
- Monique C P Mendonça
- Pharmacodelivery Group, School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| | - Ayse Kont
- Pharmacodelivery Group, School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| | - Maria Rodriguez Aburto
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, T12 XF62 Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, T12 XF62 Cork, Ireland
| | - Caitriona M O'Driscoll
- Pharmacodelivery Group, School of Pharmacy, University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
75
|
Comparison of Plasma Lipoprotein Composition and Function in Cerebral Amyloid Angiopathy and Alzheimer's Disease. Biomedicines 2021; 9:biomedicines9010072. [PMID: 33445800 PMCID: PMC7828227 DOI: 10.3390/biomedicines9010072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) refers to beta-amyloid (Aβ) deposition in brain vessels and is clinically the main cause of lobar intracerebral hemorrhage (ICH). Aβ can also accumulate in brain parenchyma forming neuritic plaques in Alzheimer’s disease (AD). Our study aimed to determine whether the peripheral lipid profile and lipoprotein composition are associated with cerebral beta-amyloidosis pathology and may reflect biological differences in AD and CAA. For this purpose, lipid and apolipoproteins levels were analyzed in plasma from 51 ICH-CAA patients (collected during the chronic phase of the disease), 60 AD patients, and 60 control subjects. Lipoproteins (VLDL, LDL, and HDL) were isolated and their composition and pro/antioxidant ability were determined. We observed that alterations in the lipid profile and lipoprotein composition were remarkable in the ICH-CAA group compared to control subjects, whereas the AD group presented no specific alterations compared with controls. ICH-CAA patients presented an atheroprotective profile, which consisted of lower total and LDL cholesterol levels. Plasma from chronic ICH-CAA patients also showed a redistribution of ApoC-III from HDL to VLDL and a higher ApoE/ApoC-III ratio in HDL. Whether these alterations reflect a protective response or have a causative effect on the pathology requires further investigation.
Collapse
|
76
|
Hu Y, Meuret C, Martinez A, Yassine HN, Nedelkov D. Distinct patterns of apolipoprotein C-I, C-II, and C-III isoforms are associated with markers of Alzheimer's disease. J Lipid Res 2020; 62:100014. [PMID: 33518512 PMCID: PMC7859854 DOI: 10.1194/jlr.ra120000919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
Apolipoproteins C-I, C-II, and C-III interact with ApoE to regulate lipoprotein metabolism and contribute to Alzheimer's disease pathophysiology. In plasma, apoC-I and C-II exist as truncated isoforms, while apoC-III exhibits multiple glycoforms. This study aimed to 1) delineate apoC-I, C-II, and C-III isoform profiles in cerebrospinal fluid (CSF) and plasma in a cohort of nondemented older individuals (n = 61), and 2) examine the effect of APOE4 on these isoforms and their correlation with CSF Aβ42, a surrogate of brain amyloid accumulation. The isoforms of the apoCs were immunoaffinity enriched and measured with MALDI-TOF mass spectrometry, revealing a significantly higher percentage of truncated apoC-I and apoC-II in CSF compared with matched plasma, with positive correlation between CSF and plasma. A greater percentage of monosialylated and disialylated apoC-III isoforms was detected in CSF, accompanied by a lower percentage of the two nonsialylated apoC-III isoforms, with significant linear correlations between CSF and plasma. Furthermore, a greater percentage of truncated apoC-I in CSF and apoC-II in plasma and CSF was observed in individuals carrying at least one APOE Ɛ4 allele. Increased apoC-I and apoC-II truncations were associated with lower CSF Aβ42. Finally, monosialylated apoC-III was lower, and disialylated apoC-III greater in the CSF of Ɛ4 carriers. Together, these results reveal distinct patterns of the apoCs isoforms in CSF, implying CSF-specific apoCs processing. These patterns were accentuated in APOE Ɛ4 allele carriers, suggesting an association between APOE4 genotype and Alzheimer's disease pathology with apoCs processing and function in the brain.
Collapse
Affiliation(s)
| | | | - Ashley Martinez
- University of Southern California, Los Angeles, California, USA
| | | | | |
Collapse
|
77
|
Pellegrini L, Albecka A, Mallery DL, Kellner MJ, Paul D, Carter AP, James LC, Lancaster MA. SARS-CoV-2 Infects the Brain Choroid Plexus and Disrupts the Blood-CSF Barrier in Human Brain Organoids. Cell Stem Cell 2020; 27:951-961.e5. [PMID: 33113348 PMCID: PMC7553118 DOI: 10.1016/j.stem.2020.10.001] [Citation(s) in RCA: 345] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 01/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, leads to respiratory symptoms that can be fatal. However, neurological symptoms have also been observed in some patients. The cause of these complications is currently unknown. Here, we use human-pluripotent-stem-cell-derived brain organoids to examine SARS-CoV-2 neurotropism. We find expression of viral receptor ACE2 in mature choroid plexus cells expressing abundant lipoproteins, but not in neurons or other cell types. We challenge organoids with SARS-CoV-2 spike pseudovirus and live virus to demonstrate viral tropism for choroid plexus epithelial cells but little to no infection of neurons or glia. We find that infected cells are apolipoprotein- and ACE2-expressing cells of the choroid plexus epithelial barrier. Finally, we show that infection with SARS-CoV-2 damages the choroid plexus epithelium, leading to leakage across this important barrier that normally prevents entry of pathogens, immune cells, and cytokines into cerebrospinal fluid and the brain.
Collapse
Affiliation(s)
- Laura Pellegrini
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Anna Albecka
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Donna L Mallery
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Max J Kellner
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - David Paul
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Andrew P Carter
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Leo C James
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
78
|
Marsillach J, Adorni MP, Zimetti F, Papotti B, Zuliani G, Cervellati C. HDL Proteome and Alzheimer's Disease: Evidence of a Link. Antioxidants (Basel) 2020; 9:E1224. [PMID: 33287338 PMCID: PMC7761753 DOI: 10.3390/antiox9121224] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Several lines of epidemiological evidence link increased levels of high-density lipoprotein-cholesterol (HDL-C) with lower risk of Alzheimer's disease (AD). This observed relationship might reflect the beneficial effects of HDL on the cardiovascular system, likely due to the implication of vascular dysregulation in AD development. The atheroprotective properties of this lipoprotein are mostly due to its proteome. In particular, apolipoprotein (Apo) A-I, E, and J and the antioxidant accessory protein paraoxonase 1 (PON1), are the main determinants of the biological function of HDL. Intriguingly, these HDL constituent proteins are also present in the brain, either from in situ expression, or derived from the periphery. Growing preclinical evidence suggests that these HDL proteins may prevent the aberrant changes in the brain that characterize AD pathogenesis. In the present review, we summarize and critically examine the current state of knowledge on the role of these atheroprotective HDL-associated proteins in AD pathogenesis and physiopathology.
Collapse
Affiliation(s)
- Judit Marsillach
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Giovanni Zuliani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| | - Carlo Cervellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| |
Collapse
|
79
|
Zhang T, Han X, Zhang X, Chen Z, Mi Y, Gou X. Dietary Fatty Acid Factors in Alzheimer's Disease: A Review. J Alzheimers Dis 2020; 78:887-904. [PMID: 33074226 DOI: 10.3233/jad-200558] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative disease characterized by brain function disorder and chronic cognitive function impairment. The onset of AD is complex and is mostly attributed to interactions between genetic factors and environmental factors. Lifestyle, dietary habits, and food consumption are likely to play indispensable functions in aged-related neurodegenerative diseases in elderly people. An increasing number of epidemiological studies have linked dietary fatty acid factors to AD, raising the point of view that fatty acid metabolism plays an important role in AD initiation and progression as well as in other central nervous system disorders. In this paper, we review the effects of the consumption of various dietary fatty acids on AD onset and progression and discuss the detrimental and beneficial effects of some typical fatty acids derived from dietary patterns on the pathology of AD. We outline these recent advances, and we recommend that healthy dietary lifestyles may contribute to preventing the occurrence and decreasing the pathology of AD.
Collapse
Affiliation(s)
- Tianying Zhang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, PR China
| | - Xiaojuan Han
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, PR China
| | - Xiaohua Zhang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, PR China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Yajing Mi
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, PR China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, PR China
| |
Collapse
|
80
|
Loupy KM, Lee T, Zambrano CA, Elsayed AI, D'Angelo HM, Fonken LK, Frank MG, Maier SF, Lowry CA. Alzheimer's Disease: Protective Effects of Mycobacterium vaccae, a Soil-Derived Mycobacterium with Anti-Inflammatory and Anti-Tubercular Properties, on the Proteomic Profiles of Plasma and Cerebrospinal Fluid in Rats. J Alzheimers Dis 2020; 78:965-987. [PMID: 33074227 DOI: 10.3233/jad-200568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is an inflammatory neurodegenerative disease that may be associated with prior bacterial infections. Microbial "old friends" can suppress exaggerated inflammation in response to disease-causing infections or increase clearance of pathogens such as Mycobacterium tuberculosis, which causes tuberculosis (TB). One such "old friend" is Mycobacterium vaccae NCTC 11659, a soil-derived bacterium that has been proposed either as a vaccine for prevention of TB, or as immunotherapy for the treatment of TB when used alongside first line anti-TB drug treatment. OBJECTIVE The goal of this study was to use a hypothesis generating approach to explore the effects of M. vaccae on physiological changes in the plasma and cerebrospinal fluid (CSF). METHODS Liquid chromatography-tandem mass spectrometry-based proteomics were performed in plasma and CSF of adult male rats after immunization with a heat-killed preparation of M. vaccae NCTC 11659 or borate-buffered saline vehicle. Gene enrichment analysis and analysis of protein-protein interactions were performed to integrate physiological network changes in plasma and CSF. We used RT-qPCR to assess immune and metabolic gene expression changes in the hippocampus. RESULTS In both plasma and CSF, immunization with M. vaccae increased proteins associated with immune activation and downregulated proteins corresponding to lipid (including phospholipid and cholesterol) metabolism. Immunization with M. vaccae also increased hippocampal expression of interleukin-4 (IL-4) mRNA, implicating anti-inflammatory effects in the central nervous system. CONCLUSION M. vaccae alters host immune activity and lipid metabolism. These data are consistent with the hypothesis that microbe-host interactions may protect against possible infection-induced, inflammation-related cognitive impairments.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Thomas Lee
- Central Analytical Laboratory and Mass Spectrometry Facility, Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Cristian A Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Heather M D'Angelo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Senior Fellow, inVIVO Planetary Health, of the Worldwide Universities Network (WUN), West New York, NJ, USA
| |
Collapse
|
81
|
Wainwright L, Hargreaves IP, Georgian AR, Turner C, Dalton RN, Abbott NJ, Heales SJR, Preston JE. CoQ 10 Deficient Endothelial Cell Culture Model for the Investigation of CoQ 10 Blood-Brain Barrier Transport. J Clin Med 2020; 9:jcm9103236. [PMID: 33050406 PMCID: PMC7601674 DOI: 10.3390/jcm9103236] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 12/31/2022] Open
Abstract
Primary coenzyme Q10 (CoQ10) deficiency is unique among mitochondrial respiratory chain disorders in that it is potentially treatable if high-dose CoQ10 supplements are given in the early stages of the disease. While supplements improve peripheral abnormalities, neurological symptoms are only partially or temporarily ameliorated. The reasons for this refractory response to CoQ10 supplementation are unclear, however, a contributory factor may be the poor transfer of CoQ10 across the blood-brain barrier (BBB). The aim of this study was to investigate mechanisms of CoQ10 transport across the BBB, using normal and pathophysiological (CoQ10 deficient) cell culture models. The study identifies lipoprotein-associated CoQ10 transcytosis in both directions across the in vitro BBB. Uptake via SR-B1 (Scavenger Receptor) and RAGE (Receptor for Advanced Glycation Endproducts), is matched by efflux via LDLR (Low Density Lipoprotein Receptor) transporters, resulting in no "net" transport across the BBB. In the CoQ10 deficient model, BBB tight junctions were disrupted and CoQ10 "net" transport to the brain side increased. The addition of anti-oxidants did not improve CoQ10 uptake to the brain side. This study is the first to generate in vitro BBB endothelial cell models of CoQ10 deficiency, and the first to identify lipoprotein-associated uptake and efflux mechanisms regulating CoQ10 distribution across the BBB. The results imply that the uptake of exogenous CoQ10 into the brain might be improved by the administration of LDLR inhibitors, or by interventions to stimulate luminal activity of SR-B1 transporters.
Collapse
Affiliation(s)
- Luke Wainwright
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK;
| | - Iain P. Hargreaves
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK;
- Department of Pharmacy and Biomolecular Science, Liverpool John Moores University, Liverpool L3 5UA, UK
| | - Ana R. Georgian
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, UK; (A.R.G.); (N.J.A.)
| | - Charles Turner
- Evelina London Children’s Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (C.T.); (R.N.D.)
| | - R. Neil Dalton
- Evelina London Children’s Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (C.T.); (R.N.D.)
| | - N. Joan Abbott
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, UK; (A.R.G.); (N.J.A.)
| | - Simon J. R. Heales
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London WC1N 3BG, UK;
- UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 6BT, UK;
| | - Jane E. Preston
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, UK; (A.R.G.); (N.J.A.)
- Correspondence: ; Tel.: +44-207-848-4881
| |
Collapse
|
82
|
Ioannou MS. Current Insights into Fatty Acid Transport in the Brain. J Membr Biol 2020; 253:375-379. [PMID: 32968835 DOI: 10.1007/s00232-020-00140-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/07/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, Canada. .,Department of Cell Biology, University of Alberta, Edmonton, Canada. .,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada. .,Group On the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Canada.
| |
Collapse
|
83
|
Tracey TJ, Kirk SE, Steyn FJ, Ngo ST. The role of lipids in the central nervous system and their pathological implications in amyotrophic lateral sclerosis. Semin Cell Dev Biol 2020; 112:69-81. [PMID: 32962914 DOI: 10.1016/j.semcdb.2020.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/11/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
Abstract
Lipids play an important role in the central nervous system (CNS). They contribute to the structural integrity and physical characteristics of cell and organelle membranes, act as bioactive signalling molecules, and are utilised as fuel sources for mitochondrial metabolism. The intricate homeostatic mechanisms underpinning lipid handling and metabolism across two major CNS cell types; neurons and astrocytes, are integral for cellular health and maintenance. Here, we explore the various roles of lipids in these two cell types. Given that changes in lipid metabolism have been identified in a number of neurodegenerative diseases, we also discuss changes in lipid handling and utilisation in the context of amyotrophic lateral sclerosis (ALS), in order to identify key cellular processes affected by the disease, and inform future areas of research.
Collapse
Affiliation(s)
- T J Tracey
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia.
| | - S E Kirk
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - F J Steyn
- Centre for Clinical Research, The University of Queensland, Brisbane, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - S T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia; Centre for Clinical Research, The University of Queensland, Brisbane, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
84
|
Lee JA, Hall B, Allsop J, Alqarni R, Allen SP. Lipid metabolism in astrocytic structure and function. Semin Cell Dev Biol 2020; 112:123-136. [PMID: 32773177 DOI: 10.1016/j.semcdb.2020.07.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/18/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Astrocytes are the most abundant glial cell in the central nervous system and are involved in multiple processes including metabolic homeostasis, blood brain barrier regulation and neuronal crosstalk. Astrocytes are the main storage point of glycogen in the brain and it is well established that astrocyte uptake of glutamate and release of lactate prevents neuronal excitability and supports neuronal metabolic function. However, the role of lipid metabolism in astrocytes in relation to neuronal support has been until recently, unclear. Lipids play a fundamental role in astrocyte function, including energy generation, membrane fluidity and cell to cell signaling. There is now emerging evidence that astrocyte storage of lipids in droplets has a crucial physiological and protective role in the central nervous system. This pathway links β-oxidation in astrocytes to inflammation, signalling, oxidative stress and mitochondrial energy generation in neurons. Disruption in lipid metabolism, structure and signalling in astrocytes can lead to pathogenic mechanisms associated with a range of neurological disorders.
Collapse
Affiliation(s)
- James Ak Lee
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Benjamin Hall
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Jessica Allsop
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Razan Alqarni
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Scott P Allen
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK.
| |
Collapse
|
85
|
Usui N, Iwata K, Miyachi T, Takagai S, Wakusawa K, Nara T, Tsuchiya KJ, Matsumoto K, Kurita D, Kameno Y, Wakuda T, Takebayashi K, Iwata Y, Fujioka T, Hirai T, Toyoshima M, Ohnishi T, Toyota T, Maekawa M, Yoshikawa T, Maekawa M, Nakamura K, Tsujii M, Sugiyama T, Mori N, Matsuzaki H. VLDL-specific increases of fatty acids in autism spectrum disorder correlate with social interaction. EBioMedicine 2020; 58:102917. [PMID: 32739868 PMCID: PMC7393524 DOI: 10.1016/j.ebiom.2020.102917] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Abnormalities of lipid metabolism contributing to the autism spectrum disorder (ASD) pathogenesis have been suggested, but the mechanisms are not fully understood. We aimed to characterize the lipid metabolism in ASD and to explore a biomarker for clinical evaluation. METHODS An age-matched case-control study was designed. Lipidomics was conducted using the plasma samples from 30 children with ASD compared to 30 typical developmental control (TD) children. Large-scale lipoprotein analyses were also conducted using the serum samples from 152 children with ASD compared to 122 TD children. Data comparing ASD to TD subjects were evaluated using univariate (Mann-Whitney test) and multivariate analyses (conditional logistic regression analysis) for main analyses using cofounders (diagnosis, sex, age, height, weight, and BMI), Spearman rank correlation coefficient, and discriminant analyses. FINDINGS Forty-eight significant metabolites involved in lipid biosynthesis and metabolism, oxidative stress, and synaptic function were identified in the plasma of ASD children by lipidomics. Among these, increased fatty acids (FAs), such as omega-3 (n-3) and omega-6 (n-6), showed correlations with clinical social interaction score and ASD diagnosis. Specific reductions of very-low-density lipoprotein (VLDL) and apoprotein B (APOB) in serum of ASD children also were found by large-scale lipoprotein analysis. VLDL-specific reduction in ASD was correlated with APOB, indicating VLDL-specific dyslipidaemia associated with APOB in ASD children. INTERPRETATION Our results demonstrated that the increases in FAs correlated positively with social interaction are due to VLDL-specific degradation, providing novel insights into the lipid metabolism underlying ASD pathophysiology. FUNDING This study was supported mainly by MEXT, Japan.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan; Center for Medical Research and Education, and Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan
| | - Taishi Miyachi
- Department of Pediatrics, Nagoya City University Medical School, Aichi 467-8601, Japan
| | - Shu Takagai
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Keisuke Wakusawa
- Department of Rehabilitation, Miyagi Children's Hospital, Miyagi 989-3126, Japan
| | - Takahiro Nara
- Department of Rehabilitation, Miyagi Children's Hospital, Miyagi 989-3126, Japan
| | - Kenji J Tsuchiya
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Kaori Matsumoto
- Graduate School of Psychology, Kanazawa Institute of Technology, Ishikawa 921-8054, Japan
| | - Daisuke Kurita
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Yosuke Kameno
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Kiyokazu Takebayashi
- Department of Psychiatry, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Yasuhide Iwata
- Department of Psychiatry and Neurology, Fukude Nishi Hospital, Shizuoka 437-1216, Japan
| | - Toru Fujioka
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan
| | - Takaharu Hirai
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Department of Community Health Nursing, School of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Manabu Toyoshima
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Tetsuo Ohnishi
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Motoko Maekawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Masato Maekawa
- Department of Laboratory Medicine, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Kazuhiko Nakamura
- Department of Psychiatry, Hirosaki University School of Medicine, Aomori 036-8562, Japan
| | - Masatsugu Tsujii
- School of Contemporary Sociology, Chukyo University, Aichi 470-0393, Japan
| | - Toshiro Sugiyama
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Norio Mori
- Department of Psychiatry and Neurology, Fukude Nishi Hospital, Shizuoka 437-1216, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka 565-0871, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan.
| |
Collapse
|
86
|
Kaiser K, Bryja V. Choroid Plexus: The Orchestrator of Long-Range Signalling Within the CNS. Int J Mol Sci 2020; 21:E4760. [PMID: 32635478 PMCID: PMC7369786 DOI: 10.3390/ijms21134760] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 01/24/2023] Open
Abstract
Cerebrospinal fluid (CSF) is the liquid that fills the brain ventricles. CSF represents not only a mechanical brain protection but also a rich source of signalling factors modulating diverse processes during brain development and adulthood. The choroid plexus (CP) is a major source of CSF and as such it has recently emerged as an important mediator of extracellular signalling within the brain. Growing interest in the CP revealed its capacity to release a broad variety of bioactive molecules that, via CSF, regulate processes across the whole central nervous system (CNS). Moreover, CP has been also recognized as a sensor, responding to altered composition of CSF associated with changes in the patterns of CNS activity. In this review, we summarize the recent advances in our understanding of the CP as a signalling centre that mediates long-range communication in the CNS. By providing a detailed account of the CP secretory repertoire, we describe how the CP contributes to the regulation of the extracellular environment-in the context of both the embryonal as well as the adult CNS. We highlight the role of the CP as an important regulator of CNS function that acts via CSF-mediated signalling. Further studies of CP-CSF signalling hold the potential to provide key insights into the biology of the CNS, with implications for better understanding and treatment of neuropathological conditions.
Collapse
Affiliation(s)
- Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| |
Collapse
|
87
|
Röhr F, Bucholtz N, Toepfer S, Norman K, Spira D, Steinhagen-Thiessen E, Lill CM, Bertram L, Demuth I, Buchmann N, Düzel S. Relationship between Lipoprotein (a) and cognitive function - Results from the Berlin Aging Study II. Sci Rep 2020; 10:10636. [PMID: 32606300 PMCID: PMC7326928 DOI: 10.1038/s41598-020-66783-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 05/26/2020] [Indexed: 12/24/2022] Open
Abstract
It has been suggested that an age-related loss of cognitive function might be driven by atherosclerotic effects associated with altered lipid patterns. However, the relationship between Lipoprotein (a) [Lp(a)] and healthy cognitive aging has not yet been sufficiently investigated. For the current analysis we used the cross-sectional data of 1,380 Berlin Aging Study II (BASE-II) participants aged 60 years and older (52.2% women, mean age 68 ± 4 years). We employed the Consortium to Establish a Registry for Alzheimer's Disease (CERAD)-Plus test battery to establish latent factors representing continuous measures of domain specific cognitive functions. Regression models adjusted for APOE genotypes, lipid parameters and other risk factors for cognitive impairment were applied to assess the association between Lp(a) and performance in specific cognitive domains. Men within the lowest Lp(a)-quintile showed better cognitive performance in the cognitive domain executive functions and processing speed (p = 0.027). No significant results were observed in women. The results of the current analysis of predominantly healthy BASE-II participants point towards an association between low Lp(a) concentrations and better cognitive performance. However, evidence for this relationship resulting from the current analysis and the employment of a differentiated cognitive assessment is rather weak.
Collapse
Affiliation(s)
- Franziska Röhr
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Nina Bucholtz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Sarah Toepfer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Kristina Norman
- German Institute of Human Nutrition, Department of Nutrition and Gerontology, Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
- Charite - Universitätsmedizin Berlin, Forschungsgruppe Geriatrie am EGZB, Berlin, Berlin, Germany
| | - Dominik Spira
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
| | - Christina M Lill
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, 23552, Lübeck, Germany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College, London, SW71, UK
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Ilja Demuth
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany.
| | - Nikolaus Buchmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany
- Department of Cardiology, Charité - University Medicine Berlin (Campus Benjamin Franklin), Berlin, Germany
| | - Sandra Düzel
- Max Planck Institute for Human Development, Berlin, Germany
| |
Collapse
|
88
|
Egyed B, Kutszegi N, Sági JC, Gézsi A, Rzepiel A, Visnovitz T, Lőrincz P, Müller J, Zombori M, Szalai C, Erdélyi DJ, Kovács GT, Semsei ÁF. MicroRNA-181a as novel liquid biopsy marker of central nervous system involvement in pediatric acute lymphoblastic leukemia. J Transl Med 2020; 18:250. [PMID: 32571344 PMCID: PMC7310470 DOI: 10.1186/s12967-020-02415-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Background Refractory central nervous system (CNS) involvement is among the major causes of therapy failure in childhood acute leukemia. Applying contemporary diagnostic methods, CNS disease is often underdiagnosed. To explore more sensitive and less invasive CNS status indicators, we examined microRNA (miR) expressions and extracellular vesicle (EV) characteristics. Methods In an acute lymphoblastic leukemia (ALL) discovery cohort, 47 miRs were screened using Custom TaqMan Advanced Low-Density Array gene expression cards. As a validation step, a candidate miR family was further scrutinized with TaqMan Advanced miRNA Assays on serial cerebrospinal fluid (CSF), bone marrow (BM) and peripheral blood samples with different acute leukemia subtypes. Furthermore, small EV-rich fractions were isolated from CSF and the samples were processed for immunoelectron microscopy with anti-CD63 and anti-CD81 antibodies, simultaneously. Results Regarding the discovery study, principal component analysis identified the role of miR-181-family (miR-181a-5p, miR-181b-5p, miR-181c-5p) in clustering CNS-positive (CNS+) and CNS-negative (CNS‒) CSF samples. We were able to validate miR-181a expression differences: it was about 52 times higher in CSF samples of CNS+ ALL patients compared to CNS‒ cases (n = 8 vs. n = 10, ΔFC = 52.30, p = 1.5E−4), and CNS+ precursor B cell subgroup also had ninefold higher miR-181a levels in their BM (p = 0.04). The sensitivity of CSF miR-181a measurement in ALL highly exceeded those of conventional cytospin in the initial diagnosis of CNS leukemia (90% vs. 54.5%). Pellet resulting from ultracentrifugation of CNS+ CSF samples of ALL patients showed atypical CD63−/CD81− small EVs in high density by immunoelectron microscopy. Conclusions After validating in extensive cohorts, quantification of miR-181a or a specific EV subtype might provide novel tools to monitor CNS disease course and further adjust CNS-directed therapy in pediatric ALL.
Collapse
Affiliation(s)
- Bálint Egyed
- 2nd Department of Pediatrics, Semmelweis University, 7-9 Tűzoltó Str, Budapest, 1094, Hungary.,Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Sqr, Budapest, 1089, Hungary
| | - Nóra Kutszegi
- 2nd Department of Pediatrics, Semmelweis University, 7-9 Tűzoltó Str, Budapest, 1094, Hungary
| | - Judit C Sági
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Sqr, Budapest, 1089, Hungary
| | - András Gézsi
- MTA-SE Immune-Proteogenomics Extracellular Vesicle Research Group, Semmelweis University, 4 Nagyvárad Sqr, Budapest, 1089, Hungary.,Department of Measurements and Information Systems, Budapest University of Technology and Economics, 2 Magyar tudosok korutja, Budapest, 1117, Hungary
| | - Andrea Rzepiel
- 2nd Department of Pediatrics, Semmelweis University, 7-9 Tűzoltó Str, Budapest, 1094, Hungary
| | - Tamás Visnovitz
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Sqr, Budapest, 1089, Hungary
| | - Péter Lőrincz
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, 1/c Pázmány Promenade, Budapest, 1117, Hungary
| | - Judit Müller
- 2nd Department of Pediatrics, Semmelweis University, 7-9 Tűzoltó Str, Budapest, 1094, Hungary
| | - Marianna Zombori
- Heim Pal National Pediatric Institute, 86 Üllői Str, Budapest, 1089, Hungary
| | - Csaba Szalai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Sqr, Budapest, 1089, Hungary.,Heim Pal National Pediatric Institute, 86 Üllői Str, Budapest, 1089, Hungary
| | - Dániel J Erdélyi
- 2nd Department of Pediatrics, Semmelweis University, 7-9 Tűzoltó Str, Budapest, 1094, Hungary
| | - Gábor T Kovács
- 2nd Department of Pediatrics, Semmelweis University, 7-9 Tűzoltó Str, Budapest, 1094, Hungary
| | - Ágnes F Semsei
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Sqr, Budapest, 1089, Hungary.
| |
Collapse
|
89
|
Chen D, Ganesh S, Wang W, Amiji M. Protein Corona-Enabled Systemic Delivery and Targeting of Nanoparticles. AAPS JOURNAL 2020; 22:83. [DOI: 10.1208/s12248-020-00464-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
|
90
|
Castaño D, Rattanasopa C, Monteiro-Cardoso VF, Corlianò M, Liu Y, Zhong S, Rusu M, Liehn EA, Singaraja RR. Lipid efflux mechanisms, relation to disease and potential therapeutic aspects. Adv Drug Deliv Rev 2020; 159:54-93. [PMID: 32423566 DOI: 10.1016/j.addr.2020.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired β-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.
Collapse
|
91
|
Mapstone M, Gross TJ, Macciardi F, Cheema AK, Petersen M, Head E, Handen BL, Klunk WE, Christian BT, Silverman W, Lott IT, Schupf N. Metabolic correlates of prevalent mild cognitive impairment and Alzheimer's disease in adults with Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12028. [PMID: 32258359 PMCID: PMC7131985 DOI: 10.1002/dad2.12028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/14/2020] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Disruption of metabolic function is a recognized feature of late onset Alzheimer's disease (LOAD). We sought to determine whether similar metabolic pathways are implicated in adults with Down syndrome (DS) who have increased risk for Alzheimer's disease (AD). METHODS We examined peripheral blood from 292 participants with DS who completed baseline assessments in the Alzheimer's Biomarkers Consortium-Down Syndrome (ABC-DS) using untargeted mass spectrometry (MS). Our sample included 38 individuals who met consensus criteria for AD (DS-AD), 43 who met criteria for mild cognitive impairment (DS-MCI), and 211 who were cognitively unaffected and stable (CS). RESULTS We measured relative abundance of 8,805 features using MS and 180 putative metabolites were differentially expressed (DE) among the groups at false discovery rate-corrected q< 0.05. From the DE features, a nine-feature classifier model classified the CS and DS-AD groups with receiver operating characteristic area under the curve (ROC AUC) of 0.86 and a two-feature model classified the DS-MCI and DS-AD groups with ROC AUC of 0.88. Metabolite set enrichment analysis across the three groups suggested alterations in fatty acid and carbohydrate metabolism. DISCUSSION Our results reveal metabolic alterations in DS-AD that are similar to those seen in LOAD. The pattern of results in this cross-sectional DS cohort suggests a dynamic time course of metabolic dysregulation which evolves with clinical progression from non-demented, to MCI, to AD. Metabolomic markers may be useful for staging progression of DS-AD.
Collapse
Affiliation(s)
- Mark Mapstone
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Thomas J Gross
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Fabio Macciardi
- Department of Psychiatry and Human BehaviorUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Amrita K Cheema
- Departments of Biochemistry and Molecular & Cellular BiologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Melissa Petersen
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Elizabeth Head
- Department of Pathology and Laboratory MedicineUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Benjamin L Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William E Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bradley T Christian
- Departments of Medical Physics and PsychiatryWaisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Wayne Silverman
- Department of PediatricsUniversity of California‐ IrvineIrvineCaliforniaUSA
| | - Ira T Lott
- Department of PediatricsUniversity of California‐ IrvineIrvineCaliforniaUSA
| | - Nicole Schupf
- Taub Institute for Research in Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of EpidemiologyJoseph P. Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | | |
Collapse
|
92
|
Hui L, Soliman ML, Geiger NH, Miller NM, Afghah Z, Lakpa KL, Chen X, Geiger JD. Acidifying Endolysosomes Prevented Low-Density Lipoprotein-Induced Amyloidogenesis. J Alzheimers Dis 2020; 67:393-410. [PMID: 30594929 DOI: 10.3233/jad-180941] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cholesterol dyshomeostasis has been linked to the pathogenesis of sporadic Alzheimer's disease (AD). In furthering the understanding of mechanisms by which increased levels of circulating cholesterol augments the risk of developing sporadic AD, others and we have reported that low-density lipoprotein (LDL) enters brain parenchyma by disrupting the blood-brain barrier and that endolysosome de-acidification plays a role in LDL-induced amyloidogenesis in neurons. Here, we tested the hypothesis that endolysosome de-acidification was central to amyloid-β (Aβ) generation and that acidifying endolysosomes protects against LDL-induced increases in Aβ levels in neurons. We demonstrated that LDL, but not HDL, de-acidified endolysosomes and increased intraneuronal and secreted levels of Aβ. ML-SA1, an agonist of endolysosome-resident TRPML1 channels, acidified endolysosomes, and TRPML1 knockdown attenuated ML-SA1-induced endolysosome acidification. ML-SA1 blocked LDL-induced increases in intraneuronal and secreted levels of Aβ as well as Aβ accumulation in endolysosomes, prevented BACE1 accumulation in endolysosomes, and decreased BACE1 activity levels. LDL downregulated TRPML1 protein levels, and TRPML1 knockdown worsens LDL-induced increases in Aβ. Our findings suggest that endolysosome acidification by activating TRPML1 may represent a protective strategy against sporadic AD.
Collapse
Affiliation(s)
- Liang Hui
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Mahmoud L Soliman
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicholas H Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicole M Miller
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Zahra Afghah
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Koffi L Lakpa
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
93
|
Meijer M, Klein M, Hannon E, van der Meer D, Hartman C, Oosterlaan J, Heslenfeld D, Hoekstra PJ, Buitelaar J, Mill J, Franke B. Genome-Wide DNA Methylation Patterns in Persistent Attention-Deficit/Hyperactivity Disorder and in Association With Impulsive and Callous Traits. Front Genet 2020; 11:16. [PMID: 32082368 PMCID: PMC7005250 DOI: 10.3389/fgene.2020.00016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/07/2020] [Indexed: 12/27/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder that often persists into adulthood. ADHD and related personality traits, such as impulsivity and callousness, are caused by genetic and environmental factors and their interplay. Epigenetic modifications of DNA, including methylation, are thought to mediate between such factors and behavior and may behave as biomarkers for disorders. Here, we set out to study DNA methylation in persistent ADHD and related traits. We performed epigenome-wide association studies (EWASs) on peripheral whole blood from participants in the NeuroIMAGE study (age range 12-23 years). We compared participants with persistent ADHD (n = 35) with healthy controls (n = 19) and with participants with remittent ADHD (n = 19). Additionally, we performed EWASs of impulsive and callous traits derived from the Conners Parent Rating Scale and the Callous-Unemotional Inventory, respectively, across all participants. For every EWAS, the linear regression model analyzed included covariates for age, sex, smoking scores, and surrogate variables reflecting blood cell type composition and genetic background. We observed no epigenome-wide significant differences in single CpG site methylation between participants with persistent ADHD and healthy controls or participants with remittent ADHD. However, epigenome-wide analysis of differentially methylated regions provided significant findings showing that hypermethylated regions in the APOB and LPAR5 genes were associated with ADHD persistence compared to ADHD remittance (p = 1.68 * 10-24 and p = 9.06 * 10-7, respectively); both genes are involved in cholesterol signaling. Both findings appeared to be linked to genetic variation in cis. We found neither significant epigenome-wide single CpG sites nor regions associated with impulsive and callous traits; the top-hits from these analyses were annotated to genes involved in neurotransmitter release and the regulation of the biological clock. No link to genetic variation was observed for these findings, which thus might reflect environmental influences. In conclusion, in this pilot study with a small sample size, we observed several DNA-methylation-disorder/trait associations of potential significance for ADHD and the related behavioral traits. Although we do not wish to draw conclusions before replication in larger, independent samples, cholesterol signaling and metabolism may be of relevance for the onset and/or persistence of ADHD.
Collapse
Affiliation(s)
- Mandy Meijer
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marieke Klein
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Eilis Hannon
- Medical School, University of Exeter, Exeter, United Kingdom
| | - Dennis van der Meer
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Faculty of Health, Medicine and Life Sciences, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Catharina Hartman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jaap Oosterlaan
- Experimental and Clinical Neuropsychology Section, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Emma Neuroscience Group, Department of Pediatrics, Amsterdam Reproduction & Development, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dirk Heslenfeld
- Experimental and Clinical Neuropsychology Section, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Pieter J. Hoekstra
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan Buitelaar
- Donders Centre for Cognitive Neuroimaging, Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
- Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, Netherlands
- Karakter Child and Adolescent Psychiatric University Centre, Nijmegen, Netherlands
| | - Jonathan Mill
- Medical School, University of Exeter, Exeter, United Kingdom
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
94
|
Ahn SI, Sei YJ, Park HJ, Kim J, Ryu Y, Choi JJ, Sung HJ, MacDonald TJ, Levey AI, Kim Y. Microengineered human blood-brain barrier platform for understanding nanoparticle transport mechanisms. Nat Commun 2020; 11:175. [PMID: 31924752 PMCID: PMC6954233 DOI: 10.1038/s41467-019-13896-7] [Citation(s) in RCA: 200] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
Challenges in drug development of neurological diseases remain mainly ascribed to the blood-brain barrier (BBB). Despite the valuable contribution of animal models to drug discovery, it remains difficult to conduct mechanistic studies on the barrier function and interactions with drugs at molecular and cellular levels. Here we present a microphysiological platform that recapitulates the key structure and function of the human BBB and enables 3D mapping of nanoparticle distributions in the vascular and perivascular regions. We demonstrate on-chip mimicry of the BBB structure and function by cellular interactions, key gene expressions, low permeability, and 3D astrocytic network with reduced reactive gliosis and polarized aquaporin-4 (AQP4) distribution. Moreover, our model precisely captures 3D nanoparticle distributions at cellular levels and demonstrates the distinct cellular uptakes and BBB penetrations through receptor-mediated transcytosis. Our BBB platform may present a complementary in vitro model to animal models for prescreening drug candidates for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Song Ih Ahn
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Yoshitaka J Sei
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hyun-Ji Park
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jinhwan Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Yujung Ryu
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jeongmoon J Choi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | | | - Allan I Levey
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - YongTae Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
95
|
APOE in the normal brain. Neurobiol Dis 2020; 136:104724. [PMID: 31911114 DOI: 10.1016/j.nbd.2019.104724] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/19/2019] [Accepted: 12/31/2019] [Indexed: 12/25/2022] Open
Abstract
The APOE4 protein affects the primary neuropathological markers of Alzheimer's disease (AD): amyloid plaques, neurofibrillary tangles, and gliosis. These interactions have been investigated to understand the strong effect of APOE genotype on risk of AD. However, APOE genotype has strong effects on processes in normal brains, in the absence of the hallmarks of AD. We propose that CNS APOE is involved in processes in the normal brains that in later years apply specifically to processes of AD pathogenesis. We review the differences of the APOE protein found in the CNS compared to the plasma, including post-translational modifications (glycosylation, lipidation, multimer formation), focusing on ways that the common APOE isoforms differ from each other. We also review structural and functional studies of young human brains and control APOE knock-in mouse brains. These approaches demonstrate the effects of APOE genotype on microscopic neuron structure, gross brain structure, and behavior, primarily related to the hippocampal areas. By focusing on the effects of APOE genotype on normal brain function, approaches can be pursued to identify biomarkers of APOE dysfunction, to promote normal functions of the APOE4 isoform, and to prevent the accumulation of the pathologic hallmarks of AD with aging.
Collapse
|
96
|
Cipollari E, Szapary HJ, Picataggi A, Billheimer JT, Lyssenko CA, Ying GS, Shaw LM, Kling MA, Kaddurah-Daouk R, Rader DJ, Pratico D, Lyssenko NN. Correlates and Predictors of Cerebrospinal Fluid Cholesterol Efflux Capacity from Neural Cells, a Family of Biomarkers for Cholesterol Epidemiology in Alzheimer's Disease. J Alzheimers Dis 2020; 74:563-578. [PMID: 32065798 PMCID: PMC7333913 DOI: 10.3233/jad-191246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Basic research has implicated intracellular cholesterol in neurons, microglia, and astrocytes in the pathogenesis of Alzheimer's disease (AD), but there is presently no assay to access intracellular cholesterol in neural cells in living people in the context of AD. OBJECTIVE To devise and characterize an assay that can access intracellular cholesterol and cholesterol efflux in neural cells in living subjects. METHODS We modified the protocol for high-density lipoprotein cholesterol efflux capacity (CEC) from macrophages, a biomarker that accesses cholesterol in macrophages in atherosclerosis. To measure cerebrospinal fluid (CSF) CECs from neurons, microglia, and astrocytes, CSF was exposed to, correspondingly, neuronal, microglial, and astrocytic cholesterol source cells. Human neuroblastoma SH-SY5Y, mouse microglial N9, and human astroglial A172 cells were used as the cholesterol source cells. CSF samples were screened for contamination with blood. CSF CECs were measured in a small cohort of 22 individuals. RESULTS CSF CECs from neurons, microglia, and astrocytes were moderately to moderately strongly correlated with CSF concentrations of cholesterol, apolipoprotein A-I, apolipoprotein E, and clusterin (Pearson's r = 0.53-0.86), were in poor agreement with one another regarding CEC of the CSF samples (Lin's concordance coefficient rc = 0.71-0.76), and were best predicted by models consisting of, correspondingly, CSF phospholipid (R2 = 0.87, p < 0.0001), CSF apolipoprotein A-I and clusterin (R2 = 0.90, p < 0.0001), and CSF clusterin (R2 = 0.62, p = 0.0005). CONCLUSION Characteristics of the CSF CEC metrics suggest a potential for independent association with AD and provision of fresh insight into the role of cholesterol in AD pathogenesis.
Collapse
Affiliation(s)
- Eleonora Cipollari
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Hannah J. Szapary
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Antonino Picataggi
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey T. Billheimer
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine A. Lyssenko
- Office of Institutional Research & Analysis, University of Pennsylvania, Philadelphia, PA, USA
| | - Gui-Shuang Ying
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchel A. Kling
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Behavioral Health Services, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Daniel J. Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Domenico Pratico
- Alzheimer’s Center at Temple, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Nicholas N. Lyssenko
- Alzheimer’s Center at Temple, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
97
|
Tanprasertsuk J, Mohn ES, Matthan NR, Lichtenstein AH, Barger K, Vishwanathan R, Johnson MA, Poon LW, Johnson EJ. Serum Carotenoids, Tocopherols, Total n-3 Polyunsaturated Fatty Acids, and n-6/n-3 Polyunsaturated Fatty Acid Ratio Reflect Brain Concentrations in a Cohort of Centenarians. J Gerontol A Biol Sci Med Sci 2019; 74:306-314. [PMID: 29893813 DOI: 10.1093/gerona/gly125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Indexed: 02/01/2023] Open
Abstract
Investigating the role of nutrition on cognitive health is challenging. Human brain tissue is inaccessible in living humans and is often limited in deceased individuals. Therefore, biomarkers of brain nutrient levels are of interest. The objective of this study was to characterize the relationships between levels of fat-soluble nutrients in serum and matched brain tissues from the frontal and temporal cortices of participants in the Georgia Centenarian Study (n = 47). After adjusting for sex, race, cognitive status (Global Deterioration Scale), body mass index, and presence of hypertension and/or diabetes, there was a significant relationship (p < 0.05) between serum and brain levels of carotenoids (lutein, zeaxanthin, cryptoxanthin, β-carotene), α-, γ-tocopherols, total n-3 polyunsaturated fatty acids (PUFAs), and n-6/n-3 PUFA ratio. The relationship between serum and brain total n-6 PUFAs was inconsistent among the two brain regions. No significant relationship was identified between serum and brain retinol, total saturated fatty acid, total monounsaturated fatty acid, and trans-fatty acid levels. These findings suggest that serum carotenoids, tocopherols, total n-3 PUFAs, and n-6/n-3 PUFA ratio reflect levels in brain and can be used as surrogate biomarkers in older population.
Collapse
Affiliation(s)
- Jirayu Tanprasertsuk
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Emily S Mohn
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Nirupa R Matthan
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Alice H Lichtenstein
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Kathryn Barger
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Rohini Vishwanathan
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Mary Ann Johnson
- Department of Foods and Nutrition, University of Georgia, Athens, Georgia.,Institute of Gerontology, University of Georgia, Athens, Georgia
| | - Leonard W Poon
- Institute of Gerontology, University of Georgia, Athens, Georgia
| | - Elizabeth J Johnson
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| |
Collapse
|
98
|
Zheng L, Fleith M, Giuffrida F, O'Neill BV, Schneider N. Dietary Polar Lipids and Cognitive Development: A Narrative Review. Adv Nutr 2019; 10:1163-1176. [PMID: 31147721 PMCID: PMC6855982 DOI: 10.1093/advances/nmz051] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Polar lipids are amphiphilic lipids with a hydrophilic head and a hydrophobic tail. Polar lipids mainly include phospholipids and sphingolipids. They are structural components of neural tissues, with the peak rate of accretion overlapping with neurodevelopmental milestones. The critical role of polar lipids in cognitive development is thought to be mediated through the regulation of signal transduction, myelination, and synaptic plasticity. Animal products (egg, meat, and dairy) are the major dietary sources of polar lipids for children and adults, whereas human milk and infant formula provide polar lipids to infants. Due to the differences observed in both concentration and proportion of polar lipids in human milk, the estimated daily intake in infants encompasses a wide range. In addition, health authorities define neither intake recommendations nor guidelines for polar lipid intake. However, adequate intake is defined for 2 nutrients that are elements of these polar lipids, namely choline and DHA. To date, limited studies exist on the brain bioavailability of dietary polar lipids via either placental transfer or the blood-brain barrier. Nevertheless, due to their role in pre- and postnatal development of the brain, there is a growing interest for the use of gangliosides, which are sphingolipids, as a dietary supplement for pregnant/lactating mothers or infants. In line with this, supplementing gangliosides and phospholipids in wild-type animals and healthy infants does suggest some positive effects on cognitive performance. Whether there is indeed added benefit of supplementing polar lipids in pregnant/lactating mothers or infants requires more clinical research. In this article, we report findings of a review of the state-of-the-art evidence on polar lipid supplementation and cognitive development. Dietary sources, recommended intake, and brain bioavailability of polar lipids are also discussed.
Collapse
Affiliation(s)
- Lu Zheng
- Nestec Ltd., Nestlé Research, Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
99
|
Essayan-Perez S, Zhou B, Nabet AM, Wernig M, Huang YWA. Modeling Alzheimer's disease with human iPS cells: advancements, lessons, and applications. Neurobiol Dis 2019; 130:104503. [PMID: 31202913 PMCID: PMC6689423 DOI: 10.1016/j.nbd.2019.104503] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/24/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
One in three people will develop Alzheimer's disease (AD) or another dementia and, despite intense research efforts, treatment options remain inadequate. Understanding the mechanisms of AD pathogenesis remains our principal hurdle to developing effective therapeutics to tackle this looming medical crisis. In light of recent discoveries from whole-genome sequencing and technical advances in humanized models, studying disease risk genes with induced human neural cells presents unprecedented advantages. Here, we first review the current knowledge of the proposed mechanisms underlying AD and focus on modern genetic insights to inform future studies. To highlight the utility of human pluripotent stem cell-based innovations, we then present an update on efforts in recapitulating the pathophysiology by induced neuronal, non-neuronal and a collection of brain cell types, departing from the neuron-centric convention. Lastly, we examine the translational potentials of such approaches, and provide our perspectives on the promise they offer to deepen our understanding of AD pathogenesis and to accelerate the development of intervention strategies for patients and risk carriers.
Collapse
Affiliation(s)
- Sofia Essayan-Perez
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Bo Zhou
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America; Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Amber M Nabet
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University Medical School, Stanford, CA 94305, United States of America
| | - Yu-Wen Alvin Huang
- Department of Molecular and Cellular Physiology, Stanford University Medical School, Stanford, CA 94305, United States of America.
| |
Collapse
|
100
|
Le Foll C. Hypothalamic Fatty Acids and Ketone Bodies Sensing and Role of FAT/CD36 in the Regulation of Food Intake. Front Physiol 2019; 10:1036. [PMID: 31474875 PMCID: PMC6702519 DOI: 10.3389/fphys.2019.01036] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022] Open
Abstract
The obesity and type-2 diabetes epidemic is escalating and represents one of the costliest biomedical challenges confronting modern society. Moreover, the increasing consumption of high fat food is often correlated with an increase in body mass index. In people predisposed to be obese or already obese, the impaired ability of the brain to monitor and respond to alterations in fatty acid (FA) metabolism is increasingly recognized as playing a role in the pathophysiological development of these disorders. The brain senses and regulates metabolism using highly specialized nutrient-sensing neurons located mainly in the hypothalamus. The same neurons are able to detect variation in the extracellular levels of glucose, FA and ketone bodies as a way to monitor nutrient availability and to alter its own activity. In addition, glial cells such as astrocytes create major connections to neurons and form a tight relationship to closely regulate nutrient uptake and metabolism. This review will examine the different pathways by which neurons are able to detect free fatty acids (FFA) to alter its activity and how high fat diet (HFD)-astrocytes induced ketone bodies production interplays with neuronal FA sensing. The role of HFD-induced inflammation and how FA modulate the reward system will also be investigated here.
Collapse
Affiliation(s)
- Christelle Le Foll
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|