1
|
Loeffler DA. Enhancing of cerebral Abeta clearance by modulation of ABC transporter expression: a review of experimental approaches. Front Aging Neurosci 2024; 16:1368200. [PMID: 38872626 PMCID: PMC11170721 DOI: 10.3389/fnagi.2024.1368200] [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: 01/10/2024] [Accepted: 05/01/2024] [Indexed: 06/15/2024] Open
Abstract
Clearance of amyloid-beta (Aβ) from the brain is impaired in both early-onset and late-onset Alzheimer's disease (AD). Mechanisms for clearing cerebral Aβ include proteolytic degradation, antibody-mediated clearance, blood brain barrier and blood cerebrospinal fluid barrier efflux, glymphatic drainage, and perivascular drainage. ATP-binding cassette (ABC) transporters are membrane efflux pumps driven by ATP hydrolysis. Their functions include maintenance of brain homeostasis by removing toxic peptides and compounds, and transport of bioactive molecules including cholesterol. Some ABC transporters contribute to lowering of cerebral Aβ. Mechanisms suggested for ABC transporter-mediated lowering of brain Aβ, in addition to exporting of Aβ across the blood brain and blood cerebrospinal fluid barriers, include apolipoprotein E lipidation, microglial activation, decreased amyloidogenic processing of amyloid precursor protein, and restricting the entrance of Aβ into the brain. The ABC transporter superfamily in humans includes 49 proteins, eight of which have been suggested to reduce cerebral Aβ levels. This review discusses experimental approaches for increasing the expression of these ABC transporters, clinical applications of these approaches, changes in the expression and/or activity of these transporters in AD and transgenic mouse models of AD, and findings in the few clinical trials which have examined the effects of these approaches in patients with AD or mild cognitive impairment. The possibility that therapeutic upregulation of ABC transporters which promote clearance of cerebral Aβ may slow the clinical progression of AD merits further consideration.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, United States
| |
Collapse
|
2
|
Han S, Yuan X, Zhao F, Manyande A, Gao F, Wang J, Zhang W, Tian X. Activation of LXRs alleviates neuropathic pain-induced cognitive dysfunction by modulation of microglia polarization and synaptic plasticity via PI3K/AKT pathway. Inflamm Res 2024; 73:157-174. [PMID: 38183431 DOI: 10.1007/s00011-023-01826-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/20/2023] [Accepted: 11/22/2023] [Indexed: 01/08/2024] Open
Abstract
OBJECTIVE Cognitive dysfunction is a common comorbidity in patients with chronic pain. Activation of Liver X receptors (LXRs) plays a potential role in improving cognitive disorders in central nervous diseases. In this study, we investigated the role of LXRs in cognitive deficits induced by neuropathic pain. METHODS We established the spared nerve injury (SNI) model to investigate pain-induced memory dysfunction. Pharmacological activation of LXRs with T0901317 or inhibition with GSK2033 was applied. PI3K inhibitor LY294002 was administered to explore the underlying mechanism of LXRs. Changes in neuroinflammation, microglia polarization, and synaptic plasticity were assessed using biochemical technologies. RESULTS We found that SNI-induced cognitive impairment was associated with reduced LXRβ expression, increased M1-phenotype microglia, decreased synaptic proteins, and inhibition of PI3K/AKT signaling pathway in the hippocampus. Activation of LXRs using T0901317 effectively alleviated SNI-induced cognitive impairment. Additionally, T0901317 promoted the polarization of microglia from M1 to M2, reduced pro-inflammatory cytokines, and upregulated synaptic proteins in the hippocampus. However, administration of GSK2033 or LY294002 abolished these protective effects of T0901317 in SNI mice. CONCLUSIONS LXRs activation alleviates neuropathic pain-induced cognitive impairment by modulating microglia polarization, neuroinflammation, and synaptic plasticity, at least partly via activation of PI3K/AKT signaling in the hippocampus. LXRs may be promising targets for addressing pain-related cognitive deficits.
Collapse
Affiliation(s)
- Siyi Han
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Xiaoman Yuan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Fengtian Zhao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Feng Gao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Jie Wang
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, Hubei Province, China
| | - Wen Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China.
| | - Xuebi Tian
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China.
| |
Collapse
|
3
|
Elder GA, Gama Sosa MA, De Gasperi R, Perez Garcia G, Perez GM, Abutarboush R, Kawoos U, Zhu CW, Janssen WGM, Stone JR, Hof PR, Cook DG, Ahlers ST. The Neurovascular Unit as a Locus of Injury in Low-Level Blast-Induced Neurotrauma. Int J Mol Sci 2024; 25:1150. [PMID: 38256223 PMCID: PMC10816929 DOI: 10.3390/ijms25021150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Blast-induced neurotrauma has received much attention over the past decade. Vascular injury occurs early following blast exposure. Indeed, in animal models that approximate human mild traumatic brain injury or subclinical blast exposure, vascular pathology can occur in the presence of a normal neuropil, suggesting that the vasculature is particularly vulnerable. Brain endothelial cells and their supporting glial and neuronal elements constitute a neurovascular unit (NVU). Blast injury disrupts gliovascular and neurovascular connections in addition to damaging endothelial cells, basal laminae, smooth muscle cells, and pericytes as well as causing extracellular matrix reorganization. Perivascular pathology becomes associated with phospho-tau accumulation and chronic perivascular inflammation. Disruption of the NVU should impact activity-dependent regulation of cerebral blood flow, blood-brain barrier permeability, and glymphatic flow. Here, we review work in an animal model of low-level blast injury that we have been studying for over a decade. We review work supporting the NVU as a locus of low-level blast injury. We integrate our findings with those from other laboratories studying similar models that collectively suggest that damage to astrocytes and other perivascular cells as well as chronic immune activation play a role in the persistent neurobehavioral changes that follow blast injury.
Collapse
Affiliation(s)
- Gregory A. Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Georgina Perez Garcia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Rania Abutarboush
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Carolyn W. Zhu
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William G. M. Janssen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James R. Stone
- Department of Radiology and Medical Imaging, University of Virginia, 480 Ray C Hunt Drive, Charlottesville, VA 22903, USA;
| | - Patrick R. Hof
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA 98108, USA;
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
| |
Collapse
|
4
|
Paseban T, Alavi MS, Etemad L, Roohbakhsh A. The role of the ATP-Binding Cassette A1 (ABCA1) in neurological disorders: a mechanistic review. Expert Opin Ther Targets 2023; 27:531-552. [PMID: 37428709 DOI: 10.1080/14728222.2023.2235718] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/09/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION Cholesterol homeostasis is critical for normal brain function. It is tightly controlled by various biological elements. ATP-binding cassette transporter A1 (ABCA1) is a membrane transporter that effluxes cholesterol from cells, particularly astrocytes, into the extracellular space. The recent studies pertaining to ABCA1's role in CNS disorders were included in this study. AREAS COVERED In this comprehensive literature review, preclinical and human studies showed that ABCA1 has a significant role in the following diseases or disorders: Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, neuropathy, anxiety, depression, psychosis, epilepsy, stroke, and brain ischemia and trauma. EXPERT OPINION ABCA1 via modulating normal and aberrant brain functions such as apoptosis, phagocytosis, BBB leakage, neuroinflammation, amyloid β efflux, myelination, synaptogenesis, neurite outgrowth, and neurotransmission promotes beneficial effects in aforementioned diseases. ABCA1 is a key molecule in the CNS. By boosting its expression or function, some CNS disorders may be resolved. In preclinical studies, liver X receptor agonists have shown promise in treating CNS disorders via ABCA1 and apoE enhancement.
Collapse
Affiliation(s)
- Tahere Paseban
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Agrawal RR, Larrea D, Xu Y, Shi L, Zirpoli H, Cummins LG, Emmanuele V, Song D, Yun TD, Macaluso FP, Min W, Kernie SG, Deckelbaum RJ, Area-Gomez E. Alzheimer's-Associated Upregulation of Mitochondria-Associated ER Membranes After Traumatic Brain Injury. Cell Mol Neurobiol 2023; 43:2219-2241. [PMID: 36571634 PMCID: PMC10287820 DOI: 10.1007/s10571-022-01299-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 10/04/2022] [Indexed: 12/27/2022]
Abstract
Traumatic brain injury (TBI) can lead to neurodegenerative diseases such as Alzheimer's disease (AD) through mechanisms that remain incompletely characterized. Similar to AD, TBI models present with cellular metabolic alterations and modulated cleavage of amyloid precursor protein (APP). Specifically, AD and TBI tissues display increases in amyloid-β as well as its precursor, the APP C-terminal fragment of 99 a.a. (C99). Our recent data in cell models of AD indicate that C99, due to its affinity for cholesterol, induces the formation of transient lipid raft domains in the ER known as mitochondria-associated endoplasmic reticulum (ER) membranes ("MAM" domains). The formation of these domains recruits and activates specific lipid metabolic enzymes that regulate cellular cholesterol trafficking and sphingolipid turnover. Increased C99 levels in AD cell models promote MAM formation and significantly modulate cellular lipid homeostasis. Here, these phenotypes were recapitulated in the controlled cortical impact (CCI) model of TBI in adult mice. Specifically, the injured cortex and hippocampus displayed significant increases in C99 and MAM activity, as measured by phospholipid synthesis, sphingomyelinase activity and cholesterol turnover. In addition, our cell type-specific lipidomics analyses revealed significant changes in microglial lipid composition that are consistent with the observed alterations in MAM-resident enzymes. Altogether, we propose that alterations in the regulation of MAM and relevant lipid metabolic pathways could contribute to the epidemiological connection between TBI and AD.
Collapse
Affiliation(s)
- Rishi R Agrawal
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA.
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| | - Delfina Larrea
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
| | - Yimeng Xu
- Biomarkers Core Laboratory, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 10-105, New York, NY, 10032, USA
| | - Lingyan Shi
- Department of Chemistry, Columbia University, 3000 Broadway, Havemeyer Hall, New York, NY, 10027, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Hylde Zirpoli
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA
| | - Leslie G Cummins
- Analytical Imaging Facility, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, 10461, USA
| | - Valentina Emmanuele
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
| | - Donghui Song
- Department of Chemistry, Columbia University, 3000 Broadway, Havemeyer Hall, New York, NY, 10027, USA
| | - Taekyung D Yun
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
| | - Frank P Macaluso
- Analytical Imaging Facility, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, 10461, USA
| | - Wei Min
- Biomarkers Core Laboratory, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 10-105, New York, NY, 10032, USA
| | - Steven G Kernie
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
- Department of Pediatrics, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 17, New York, NY, 10032, USA
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA
- Department of Pediatrics, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 17, New York, NY, 10032, USA
| | - Estela Area-Gomez
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA.
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA.
- Centro de Investigaciones Biológicas Margarita Salas - CSIC, C. Ramiro de Maeztu, 9, 28040, Madrid, Spain.
| |
Collapse
|
6
|
Shao F, Wang X, Wu H, Wu Q, Zhang J. Microglia and Neuroinflammation: Crucial Pathological Mechanisms in Traumatic Brain Injury-Induced Neurodegeneration. Front Aging Neurosci 2022; 14:825086. [PMID: 35401152 PMCID: PMC8990307 DOI: 10.3389/fnagi.2022.825086] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most common diseases in the central nervous system (CNS) with high mortality and morbidity. Patients with TBI usually suffer many sequelae in the life time post injury, including neurodegenerative disorders such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). However, the pathological mechanisms connecting these two processes have not yet been fully elucidated. It is important to further investigate the pathophysiological mechanisms underlying TBI and TBI-induced neurodegeneration, which will promote the development of precise treatment target for these notorious neurodegenerative consequences after TBI. A growing body of evidence shows that neuroinflammation is a pivotal pathological process underlying chronic neurodegeneration following TBI. Microglia, as the immune cells in the CNS, play crucial roles in neuroinflammation and many other CNS diseases. Of interest, microglial activation and functional alteration has been proposed as key mediators in the evolution of chronic neurodegenerative pathology following TBI. Here, we review the updated studies involving phenotypical and functional alterations of microglia in neurodegeneration after injury, survey key molecules regulating the activities and functional responses of microglia in TBI pathology, and explore their potential implications to chronic neurodegeneration after injury. The work will give us a comprehensive understanding of mechanisms driving TBI-related neurodegeneration and offer novel ideas of developing corresponding prevention and treatment strategies for this disease.
Collapse
Affiliation(s)
- Fangjie Shao
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qun Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Qun Wu,
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
- Jianmin Zhang,
| |
Collapse
|
7
|
Srinivasan G, Brafman DA. The Emergence of Model Systems to Investigate the Link Between Traumatic Brain Injury and Alzheimer's Disease. Front Aging Neurosci 2022; 13:813544. [PMID: 35211003 PMCID: PMC8862182 DOI: 10.3389/fnagi.2021.813544] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous epidemiological studies have demonstrated that individuals who have sustained a traumatic brain injury (TBI) have an elevated risk for developing Alzheimer's disease and Alzheimer's-related dementias (AD/ADRD). Despite these connections, the underlying mechanisms by which TBI induces AD-related pathology, neuronal dysfunction, and cognitive decline have yet to be elucidated. In this review, we will discuss the various in vivo and in vitro models that are being employed to provide more definite mechanistic relationships between TBI-induced mechanical injury and AD-related phenotypes. In particular, we will highlight the strengths and weaknesses of each of these model systems as it relates to advancing the understanding of the mechanisms that lead to TBI-induced AD onset and progression as well as providing platforms to evaluate potential therapies. Finally, we will discuss how emerging methods including the use of human induced pluripotent stem cell (hiPSC)-derived cultures and genome engineering technologies can be employed to generate better models of TBI-induced AD.
Collapse
Affiliation(s)
| | - David A. Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
8
|
Barrett JP, Knoblach SM, Bhattacharya S, Gordish-Dressman H, Stoica BA, Loane DJ. Traumatic Brain Injury Induces cGAS Activation and Type I Interferon Signaling in Aged Mice. Front Immunol 2021; 12:710608. [PMID: 34504493 PMCID: PMC8423402 DOI: 10.3389/fimmu.2021.710608] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/05/2021] [Indexed: 12/21/2022] Open
Abstract
Aging adversely affects inflammatory processes in the brain, which has important implications in the progression of neurodegenerative disease. Following traumatic brain injury (TBI), aged animals exhibit worsened neurological function and exacerbated microglial-associated neuroinflammation. Type I Interferons (IFN-I) contribute to the development of TBI neuropathology. Further, the Cyclic GMP-AMP Synthase (cGAS) and Stimulator of Interferon Genes (STING) pathway, a key inducer of IFN-I responses, has been implicated in neuroinflammatory activity in several age-related neurodegenerative diseases. Here, we set out to investigate the effects of TBI on cGAS/STING activation, IFN-I signaling and neuroinflammation in young and aged C57Bl/6 male mice. Using a controlled cortical impact model, we evaluated transcriptomic changes in the injured cortex at 24 hours post-injury, and confirmed activation of key neuroinflammatory pathways in biochemical studies. TBI induced changes were highly enriched for transcripts that were involved in inflammatory responses to stress and host defense. Deeper analysis revealed that TBI increased expression of IFN-I related genes (e.g. Ifnb1, Irf7, Ifi204, Isg15) and IFN-I signaling in the injured cortex of aged compared to young mice. There was also a significant age-related increase in the activation of the DNA-recognition pathway, cGAS, which is a key mechanism to propagate IFN-I responses. Finally, enhanced IFN-I signaling in the aged TBI brain was confirmed by increased phosphorylation of STAT1, an important IFN-I effector molecule. This age-related activation of cGAS and IFN-I signaling may prove to be a mechanistic link between microglial-associated neuroinflammation and neurodegeneration in the aged TBI brain.
Collapse
Affiliation(s)
- James P Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Susan M Knoblach
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Health System, Washington, DC, United States.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Surajit Bhattacharya
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Health System, Washington, DC, United States
| | - Heather Gordish-Dressman
- Center for Translational Science, Children's Research Institute, Children's National Health System, Washington, DC, United States.,Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, United States.,Veterans Affairs (VA) Maryland Health Care System, Baltimore VA Medical Center, Baltimore, MD, United States
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, United States.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
9
|
Perez Garcia G, De Gasperi R, Tschiffely AE, Gama Sosa MA, Abutarboush R, Kawoos U, Statz JK, Ciarlone S, Reed EM, Jeyarajah T, Perez G, Otero Pagan A, Pryor D, Hof P, Cook D, Gandy S, Elder G, Ahlers S. Repetitive low-level blast exposure improves behavioral deficits and chronically lowers Aβ42 in an Alzheimer's disease transgenic mouse model. J Neurotrauma 2021; 38:3146-3173. [PMID: 34353119 DOI: 10.1089/neu.2021.0184] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Public awareness of traumatic brain injury (TBI) in the military increased recently because of the conflicts in Iraq and Afghanistan where blast injury was the most common mechanism of injury. Besides overt injuries, concerns also exist over the potential adverse consequences of subclinical blast exposures, which are common for many service members. TBI is a risk factor for the later development of neurodegenerative diseases, including Alzheimer's disease (AD)-like disorders. Studies of acute TBI in humans and animals have suggested that increased processing of the amyloid precursor protein (APP) towards the amyloid beta protein (Aβ) may explain the epidemiological associations with AD. However, in a prior study we found in both rat and mouse models of blast overpressure exposure (BOP), that rather than increasing, rodent brain Aβ42 levels were decreased following acute blast exposure. Here we subjected APP/presenilin 1 transgenic mice (APP/PS1 Tg) to an extended sequence of repetitive low-level blast exposures (34.5 kPa) administered three times per week over 8 weeks. If initiated at 20 weeks of age, these repetitive exposures, which were designed to mimic human subclinical blast exposures, reduced anxiety and improved cognition as well as social interactions in APP/PS1 Tg mice, returning many behavioral parameters in APP/PS1 Tg mice to levels of non-transgenic wild type mice. Repetitive low-level blast exposure was less effective at improving behavioral deficits in APP/PS1 Tg mice when begun at 36 weeks of age. While amyloid plaque loads were unchanged, Aβ42 levels and Aβ oligomers were reduced in brain of mice exposed to repetitive low-level blast exposures initiated at 20 weeks of age, although levels did not directly correlate with behavioral parameters in individual animals. These results have implications for understanding the nature of blast effects on the brain and their relationship to human neurodegenerative diseases.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Icahn School of Medicine at Mount Sinai, 5925, Neurology, 1468 Madison Avenue Annenberg Building Floor 14 Room 60, New York, New York, New York, United States, 10029-6574.,James J Peters VA Medical Center, 20071, Research, 130 W Kingsbridge Rd, The Bronx, NY 10468, Bronx, United States, 10468-3904;
| | - Rita De Gasperi
- James J. Peters VA Medical Center, Research and Development, 130 west kingsbridge road, RD 3F-20, Bronx, New York, United States, 10468;
| | - Anna E Tschiffely
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Miguel A Gama Sosa
- James J. Peters VA Medical Center, Research and Development, 130 W Kingsbridge Rd, Bronx, New York, United States, 10468;
| | - Rania Abutarboush
- Naval Medical Research Center, 19930, Neurotrauma, 503 Robert Grant Ave, Silver Spring, Maryland, United States, 20910;
| | - Usmah Kawoos
- Naval Medical Research Center, 19930, Neurotrauma, 503 Robert Grant Ave, Silver Spring, Maryland, United States, 20910.,Henry M Jackson Foundation for the Advancement of Military Medicine Inc, 44069, Bethesda, Maryland, United States;
| | | | - Stephanie Ciarlone
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Eileen M Reed
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Theepica Jeyarajah
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Gissel Perez
- James J Peters VA Medical Center, 20071, Research and Development, Bronx, New York, United States;
| | - Alena Otero Pagan
- James J Peters VA Medical Center, 20071, Research and Development, Bronx, New York, United States;
| | - Dylan Pryor
- James J Peters VA Medical Center, 20071, Research, 130 W. Kingsbridge Rd., Bronx, New York, United States, 10468;
| | - Patrick Hof
- Icahn School of Medicine at Mount Sinai, 5925, New York, New York, United States;
| | - David Cook
- VA Puget Sound Health Care System, 20128, Geriatric Research, Education, and Clinical Center, 1660 S Columbian Way, Seattle, Washington, United States, 98108.,University of Washington, 7284, Division of Gerontology and Geriatric Medicine, Seattle, Washington, United States;
| | - Samuel Gandy
- 88 Mercer AvenueHartsdaleHartsdale, New York, United States, 10530.,Sam Gandy, 88 Mercer Avenue, United States;
| | - Gregory Elder
- James J. Peters VAMC, Research and Development 3F22, 130 West Kingsbridge Road, Bronx, New York, United States, 10468;
| | - Stephen Ahlers
- Naval Medical Research Center, OUMD, 503 Robert Grant Ave, Silver Spring, Maryland, United States, 20910;
| |
Collapse
|
10
|
Jacobo-Albavera L, Domínguez-Pérez M, Medina-Leyte DJ, González-Garrido A, Villarreal-Molina T. The Role of the ATP-Binding Cassette A1 (ABCA1) in Human Disease. Int J Mol Sci 2021; 22:ijms22041593. [PMID: 33562440 PMCID: PMC7915494 DOI: 10.3390/ijms22041593] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Cholesterol homeostasis is essential in normal physiology of all cells. One of several proteins involved in cholesterol homeostasis is the ATP-binding cassette transporter A1 (ABCA1), a transmembrane protein widely expressed in many tissues. One of its main functions is the efflux of intracellular free cholesterol and phospholipids across the plasma membrane to combine with apolipoproteins, mainly apolipoprotein A-I (Apo A-I), forming nascent high-density lipoprotein-cholesterol (HDL-C) particles, the first step of reverse cholesterol transport (RCT). In addition, ABCA1 regulates cholesterol and phospholipid content in the plasma membrane affecting lipid rafts, microparticle (MP) formation and cell signaling. Thus, it is not surprising that impaired ABCA1 function and altered cholesterol homeostasis may affect many different organs and is involved in the pathophysiology of a broad array of diseases. This review describes evidence obtained from animal models, human studies and genetic variation explaining how ABCA1 is involved in dyslipidemia, coronary heart disease (CHD), type 2 diabetes (T2D), thrombosis, neurological disorders, age-related macular degeneration (AMD), glaucoma, viral infections and in cancer progression.
Collapse
Affiliation(s)
- Leonor Jacobo-Albavera
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
| | - Mayra Domínguez-Pérez
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
| | - Diana Jhoseline Medina-Leyte
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Mexico City CP04510, Mexico
| | - Antonia González-Garrido
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
| | - Teresa Villarreal-Molina
- Laboratorio de Genómica de Enfermedades Cardiovasculares, Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City CP14610, Mexico; (L.J.-A.); (M.D.-P.); (D.J.M.-L.); (A.G.-G.)
- Correspondence:
| |
Collapse
|
11
|
Abrahamson EE, Ikonomovic MD. Brain injury-induced dysfunction of the blood brain barrier as a risk for dementia. Exp Neurol 2020; 328:113257. [PMID: 32092298 DOI: 10.1016/j.expneurol.2020.113257] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic physiological interface between brain parenchyma and cerebral vasculature. It is composed of closely interacting cells and signaling molecules that regulate movement of solutes, ions, nutrients, macromolecules, and immune cells into the brain and removal of products of normal and abnormal brain cell metabolism. Dysfunction of multiple components of the BBB occurs in aging, inflammatory diseases, traumatic brain injury (TBI, severe or mild repetitive), and in chronic degenerative dementing disorders for which aging, inflammation, and TBI are considered risk factors. BBB permeability changes after TBI result in leakage of serum proteins, influx of immune cells, perivascular inflammation, as well as impairment of efflux transporter systems and accumulation of aggregation-prone molecules involved in hallmark pathologies of neurodegenerative diseases with dementia. In addition, cerebral vascular dysfunction with persistent alterations in cerebral blood flow and neurovascular coupling contribute to brain ischemia, neuronal degeneration, and synaptic dysfunction. While the idea of TBI as a risk factor for dementia is supported by many shared pathological features, it remains a hypothesis that needs further testing in experimental models and in human studies. The current review focusses on pathological mechanisms shared between TBI and neurodegenerative disorders characterized by accumulation of pathological protein aggregates, such as Alzheimer's disease and chronic traumatic encephalopathy. We discuss critical knowledge gaps in the field that need to be explored to clarify the relationship between TBI and risk for dementia and emphasize the need for longitudinal in vivo studies using imaging and biomarkers of BBB dysfunction in people with single or multiple TBI.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
12
|
Ojo JO, Leary P, Lungmus C, Algamal M, Mouzon B, Bachmeier C, Mullan M, Stewart W, Crawford F. Subchronic Pathobiological Response Following Chronic Repetitive Mild Traumatic Brain Injury in an Aged Preclinical Model of Amyloid Pathogenesis. J Neuropathol Exp Neurol 2019; 77:1144-1162. [PMID: 30395237 DOI: 10.1093/jnen/nly101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
Repetitive mild traumatic brain injury (r-mTBI) is a risk factor for Alzheimer disease (AD). The precise nature of how r-mTBI leads to, or precipitates, AD pathogenesis remains unclear. In this study, we explore subchronic effects of chronic r-mTBI (12-impacts) administered over 1-month in aged-PS1/APP mice and littermate controls. We investigate specific mechanisms that may elucidate the molecular link between AD and r-mTBI, focusing primarily on amyloid and tau pathology, amyloid processing, glial activation states, and associated clearance mechanisms. Herein, we demonstrate r-mTBI in aged PS1/APP mice does not augment, glial activation, amyloid burden, or tau pathology (with exception of pS202-positive Tau) 1 month after exposure to the last-injury. However, we observed a decrease in brain soluble Aβ42 levels without any appreciable change in peripheral soluble Aβ42 levels. This was accompanied by an increase in brain insoluble to soluble Aβ42 ratio in injured PS1/APP mice compared with sham injury. A parallel reduction in phagocytic receptor, triggering receptor expressed on myeloid cells 2, was also observed. This study demonstrates very subtle subchronic effects of r-mTBI on a preexisting amyloid pathology background, which may be on a continuum toward a slow and worsening neurodegenerative outcome compared with sham injury, and therefore, have many implications, especially in the elderly population exposed to TBI.
Collapse
Affiliation(s)
- Joseph O Ojo
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,James A. Haley Veterans' Hospital, Tampa, Florida.,Open University, Milton Keynes, UK
| | - Paige Leary
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida
| | - Caryln Lungmus
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida
| | - Moustafa Algamal
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,Open University, Milton Keynes, UK
| | - Benoit Mouzon
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,James A. Haley Veterans' Hospital, Tampa, Florida.,Open University, Milton Keynes, UK
| | - Corbin Bachmeier
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,Open University, Milton Keynes, UK.,Bay Pines VA Healthcare System, Bay Pines, Florida
| | - Michael Mullan
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,Open University, Milton Keynes, UK
| | - William Stewart
- Queen Elizabeth University Hospital and University of Glasgow, Glasgow, UK.,University of Pennsylvania, Philadelphia, Pennsylvania
| | - Fiona Crawford
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,James A. Haley Veterans' Hospital, Tampa, Florida.,Open University, Milton Keynes, UK
| |
Collapse
|
13
|
McCarty MF, O'Keefe JH, DiNicolantonio JJ. A diet rich in taurine, cysteine, folate, B 12 and betaine may lessen risk for Alzheimer's disease by boosting brain synthesis of hydrogen sulfide. Med Hypotheses 2019; 132:109356. [PMID: 31450076 DOI: 10.1016/j.mehy.2019.109356] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/06/2019] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
The gaseous physiological modulator hydrogen sulfide (H2S) has recently been shown to exert a variety of neuroprotective effects. In particular, the treatment of transgenic mouse models of Alzheimer's disease (AD) with agents that release H2S aids preservation of cognitive function, suppresses brain production of amyloid beta, and decreases tau phosphorylation. The possible physiological relevance of these findings is suggested by the finding that brain and plasma levels of H2S are markedly lower in AD patients than matched controls. Hence, nutraceutical strategies which boost brain synthesis or levels of H2S may have potential for prevention of AD. The chief enzyme which synthesizes H2S in brain parenchyma, cystathionine beta-synthase (CBS), employs cysteine as its rate-limiting substrate, and is allosterically activated by S-adenosylmethionine (SAM). Supplemental taurine has been shown to boost expression of this enzyme, as well as that of another H2S source, cystathionine gamma-lyase, in vascular tissue, and to enhance plasma H2S levels; in rats subjected to hemorrhagic stroke, co-administration of taurine has been shown to blunt a marked reduction in brain CBS expression. Brain levels of SAM are about half as high in AD patients as in controls, and this is thought to explain the reduction of brain H2S in these patients. These considerations suggest that supplementation with cysteine, taurine, and agents which promote methyl group availability - such as SAM, folate, vitamin B12, and betaine - may have potential for boosting brain synthesis of H2S and thereby aiding AD prevention. Indeed, most of these agents have already demonstrated utility in mouse AD models - albeit the extent to which increased H2S synthesis contributes to this protection remains unclear. Moreover, prospective epidemiology has associated low dietary or plasma levels of folate, B12, and taurine with increased dementia risk. Rodent studies suggest that effective nutraceutical strategies for boosting brain H2S synthesis may in fact have broad neuroprotective utility, possibly aiding prevention and/or control not only of AD but also Parkinson's disease and glaucoma, while diminishing the neuronal damage associated with brain trauma or stroke.
Collapse
Affiliation(s)
| | - James H O'Keefe
- Saint Luke's Mid America Heart Institute, Kansas City, MO, United States
| | | |
Collapse
|
14
|
Targeting Apolipoprotein E for Alzheimer's Disease: An Industry Perspective. Int J Mol Sci 2019; 20:ijms20092161. [PMID: 31052389 PMCID: PMC6539182 DOI: 10.3390/ijms20092161] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/27/2019] [Accepted: 04/28/2019] [Indexed: 02/08/2023] Open
Abstract
Apolipoprotein E (apoE), a key lipid transport protein in the brain, is predominantly produced by astrocytes. Astrocytes are the most numerous cell type in the brain and are the main support network for neurons. They play a critical role in the synthesis and delivery of cholesterol in the brain. Humans have three common apoE isoforms, apoE2, apoE3 and apoE4, that show a strong genotype effect on the risk and age of onset for sporadic and late onset forms of Alzheimer’s disease (AD). Carriers of an ε4 allele have an increased risk of developing AD, while those with an ε2 allele are protected. Investigations into the contribution of apoE to the development of AD has yielded conflicting results and there is still much speculation about the role of this protein in disease. Here, we review the opposing hypotheses currently described in the literature and the approaches that have been considered for targeting apoE as a novel therapeutic strategy for AD. Additionally, we provide our perspective on the rationale for targeting apoE and the challenges that arise with respect to “drug-ability” of this target.
Collapse
|
15
|
Xu GB, Yang LQ, Guan PP, Wang ZY, Wang P. Prostaglandin A1 Inhibits the Cognitive Decline of APP/PS1 Transgenic Mice via PPARγ/ABCA1-dependent Cholesterol Efflux Mechanisms. Neurotherapeutics 2019; 16:505-522. [PMID: 30627958 PMCID: PMC6554490 DOI: 10.1007/s13311-018-00704-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Prostaglandins (PGs) are early and key contributors to chronic neurodegenerative diseases. As one important member of classical PGs, PGA1 has been reported to exert potential neuroprotective effects. However, the mechanisms remain unknown. To this end, we are prompted to investigate whether PGA1 is a useful neurological treatment for Alzheimer's disease (AD) or not. Using high-throughput sequencing, we found that PGA1 potentially regulates cholesterol metabolism and lipid transport. Interestingly, we further found that short-term administration of PGA1 decreased the levels of the monomeric and oligomeric β-amyloid protein (oAβ) in a cholesterol-dependent manner. In detail, PGA1 activated the peroxisome proliferator-activated receptor-gamma (PPARγ) and ATP-binding cassette subfamily A member 1 (ABCA1) signalling pathways, promoting the efflux of cholesterol and decreasing the intracellular cholesterol levels. Through PPARγ/ABCA1/cholesterol-dependent pathway, PGA1 decreased the expression of presenilin enhancer protein 2 (PEN-2), which is responsible for the production of Aβ. More importantly, long-term administration of PGA1 remarkably decreased the formation of Aβ monomers, oligomers, and fibrils. The actions of PGA1 on the production and deposition of Aβ ultimately improved the cognitive decline of the amyloid precursor protein/presenilin1 (APP/PS1) transgenic mice.
Collapse
Affiliation(s)
- Guo-Biao Xu
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China
| | - Liu-Qing Yang
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China
| | - Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China.
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11. Wenhua Road, Shenyang, 110819, People's Republic of China.
| |
Collapse
|
16
|
Hagos FT, Adams SM, Poloyac SM, Kochanek PM, Horvat CM, Clark RSB, Empey PE. Membrane transporters in traumatic brain injury: Pathological, pharmacotherapeutic, and developmental implications. Exp Neurol 2019; 317:10-21. [PMID: 30797827 DOI: 10.1016/j.expneurol.2019.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
Membrane transporters regulate the trafficking of endogenous and exogenous molecules across biological barriers and within the neurovascular unit. In traumatic brain injury (TBI), they moderate the dynamic movement of therapeutic drugs and injury mediators among neurons, endothelial cells and glial cells, thereby becoming important determinants of pathogenesis and effective pharmacotherapy after TBI. There are three ways transporters may impact outcomes in TBI. First, transporters likely play a key role in the clearance of injury mediators. Second, genetic association studies suggest transporters may be important in the transition of TBI from acute brain injury to a chronic neurological disease. Third, transporters dynamically control the brain penetration and efflux of many drugs and their distribution within and elimination from the brain, contributing to pharmacoresistance and possibly in some cases pharmacosensitivity. Understanding the nature of drugs or candidate drugs in development with respect to whether they are a transporter substrate or inhibitor is relevant to understand whether they distribute to their target in sufficient concentrations. Emerging data provide evidence of altered expression and function of transporters in humans after TBI. Genetic variability in expression and/or function of key transporters adds an additional dynamic, as shown in recent clinical studies. In this review, evidence supporting the role of individual membrane transporters in TBI are discussed as well as novel strategies for their modulation as possible therapeutic targets. Since data specifically targeting pediatric TBI are sparse, this review relies mainly on experimental studies using adult animals and clinical studies in adult patients.
Collapse
Affiliation(s)
- Fanuel T Hagos
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America
| | - Solomon M Adams
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America
| | - Samuel M Poloyac
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States of America
| | - Christopher M Horvat
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States of America
| | - Robert S B Clark
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States of America.
| | - Philip E Empey
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
17
|
Anthony Jalin AMA, Jin R, Wang M, Li G. EPPS treatment attenuates traumatic brain injury in mice by reducing Aβ burden and ameliorating neuronal autophagic flux. Exp Neurol 2019; 314:20-33. [PMID: 30639321 DOI: 10.1016/j.expneurol.2019.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
Abstract
Beta-amyloid (Aβ) burden and impaired neuronal autophagy contribute to secondary brain injury after traumatic brain injury (TBI). 4-(2-hydroxyethyl)-1-piperazinepropanesulphonic acid (EPPS) treatment has been reported to reduce Aβ aggregation and rescue behavioral deficits in Alzheimer's disease-like mice. Here, we investigated neuroprotective effects of EPPS in a mouse model of TBI. Mice subjected to controlled cortical impact (CCI) were treated with EPPS (120 mg/kg, orally) immediately after CCI and thereafter once daily for 3 or 7 days. We found that EPPS treatment profoundly reduced the accumulation of beta-amyloid precursor protein (β-APP) and Aβ over a widespread area detected in the pericontusional cortex, external capsule (EC), and hippocampal CA1 and CA3 at 3 days after TBI, accompanied by significant reduction in the TBI-induced diffuse axonal injury identified by increased immunoreactivity of SMI-32 (an indicator for axonal damage). We also found that EPPS treatment ameliorated the TBI-induced synaptic damage (as reflected by enhanced postsynaptic density 95, PSD-95), and impairment of autophagy flux in the neurons as reflected by reduced autophagy markers (LC3-II/LC3-I ratio and p62/SQSTM1) and increased lysosomal enzyme cathepsin D (CTSD) in neurons detected in the cortex and hippocampal CA1. As a result, EPPS treatment significantly reduced the TBI-induced early neuronal apoptosis (assessed by active caspase-3), and eventually prevented cortical tissue loss and hippocampal neuronal loss at 28 days after TBI. Additionally, we found that inhibition of autophagic flux with chloroquine by decreasing autophagosome-lysosome fusion significantly reversed the decreased expressions of neuronal p62/SQSTM1 and apoptosis by EPPS treatment. These data suggest that the neuroprotection by EPPS is, at least in part, related to improved autophagy flux. Finally, we found that EPPS treatment significantly improved the cortex-dependent motor and hippocampal-dependent cognitive deficits associated with TBI. Taken together, these findings support the further investigation of EPPS as a treatment for TBI.
Collapse
Affiliation(s)
| | - Rong Jin
- Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey 17033, USA.
| | - Min Wang
- Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey 17033, USA.
| | - Guohong Li
- Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey 17033, USA.
| |
Collapse
|
18
|
Petty HR. Frontiers of Complex Disease Mechanisms: Membrane Surface Tension May Link Genotype to Phenotype in Glaucoma. Front Cell Dev Biol 2018; 6:32. [PMID: 29682502 PMCID: PMC5897435 DOI: 10.3389/fcell.2018.00032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/13/2018] [Indexed: 12/19/2022] Open
Abstract
Although many monogenic diseases are understood based upon structural changes of gene products, less progress has been made concerning polygenic disease mechanisms. This article presents a new interdisciplinary approach to understand complex diseases, especially their genetic polymorphisms. I focus upon primary open angle glaucoma (POAG). Although elevated intraocular pressure (IOP) and oxidative stress are glaucoma hallmarks, the linkages between these factors and cell death are obscure. Reactive oxygen species (ROS) promote the formation of oxidatively truncated phosphoglycerides (OTP), free fatty acids, lysophosphoglycerides, oxysterols, and other chemical species that promote membrane disruption and decrease membrane surface tension. Several POAG-linked gene polymorphisms identify proteins that manage damaged lipids and/or influence membrane surface tension. POAG-related genes expected to participate in these processes include: ELOVL5, ABCA1, APOE4, GST, CYP46A1, MYOC, and CAV. POAG-related gene products are expected to influence membrane surface tension, strength, and repair. I propose that heightened IOP overcomes retinal ganglion cell (RGC) membrane compressive strength, weakened by damaged lipid accumulation, to form pores. The ensuing structural failure promotes apoptosis and blindness. The linkage between glaucoma genotype and phenotype is mediated by physical events. Force balancing between the IOP and compressive strength regulates pore nucleation; force balancing between pore line tension and membrane surface tension regulates pore growth. Similar events may contribute to traumatic brain injury, Alzheimer's disease, and macular degeneration.
Collapse
Affiliation(s)
- Howard R Petty
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
19
|
Main BS, Villapol S, Sloley SS, Barton DJ, Parsadanian M, Agbaegbu C, Stefos K, McCann MS, Washington PM, Rodriguez OC, Burns MP. Apolipoprotein E4 impairs spontaneous blood brain barrier repair following traumatic brain injury. Mol Neurodegener 2018; 13:17. [PMID: 29618365 PMCID: PMC5885297 DOI: 10.1186/s13024-018-0249-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Traumatic Brain Injury (TBI) is a major cause of disability and mortality, to which there is currently no comprehensive treatment. Blood Brain Barrier (BBB) dysfunction is well documented in human TBI patients, yet the molecular mechanisms that underlie this neurovascular unit (NVU) pathology remains unclear. The apolipoprotein-E (apoE) protein has been implicated in controlling BBB integrity in an isoform dependent manner, via suppression of Cyclophilin A (CypA)-Matrix metallopeptidase-9 (MMP-9) signaling cascades, however the contribution of this pathway in TBI-induced BBB permeability is not fully investigated. METHODS We exposed C57Bl/6 mice to controlled cortical impact and assessed NVU and BBB permeability responses up to 21 days post-injury. We pharmacologically probed the role of the CypA-MMP-9 pathway in BBB permeability after TBI using Cyclosporin A (CsA, 20 mg/kg). Finally, as the apoE4 protein is known to be functionally deficient compared to the apoE3 protein, we used humanized APOE mice as a clinically relevant model to study the role of apoE on BBB injury and repair after TBI. RESULTS In C57Bl/6 mice there was an inverse relationship between soluble apoE and BBB permeability, such that damaged BBB stabilizes as apoE levels increase in the days following TBI. TBI mice displayed acute pericyte loss, increased MMP-9 production and activity, and reduced tight-junction expression. Treatment with the CypA antagonist CsA in C57Bl/6 mice attenuates MMP-9 responses and enhances BBB repair after injury, demonstrating that MMP-9 plays an important role in the timing of spontaneous BBB repair after TBI. We also show that apoe mRNA is present in both astrocytes and pericytes after TBI. We report that APOE3 and APOE4 mice have similar acute BBB responses to TBI, but APOE3 mice display faster spontaneous BBB repair than APOE4 mice. Isolated microvessel analysis reveals delayed pericyte repopulation, augmented and sustained MMP-9 expression at the NVU, and impaired stabilization of Zonula Occludens-1, Occludin and Claudin-5 expression at tight junctions in APOE4 mice after TBI compared to APOE3 mice. CONCLUSIONS These data confirm apoE as an important modulator of spontaneous BBB stabilization following TBI, and highlights the APOE4 allele as a risk factor for poor outcome after TBI.
Collapse
Affiliation(s)
- Bevan S Main
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Sonia Villapol
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Stephanie S Sloley
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - David J Barton
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Maia Parsadanian
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Chinyere Agbaegbu
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Kathryn Stefos
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Mondona S McCann
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Patricia M Washington
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Olga C Rodriguez
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Mark P Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, 20057, USA. .,Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, D.C, 20057, USA.
| |
Collapse
|
20
|
The protective effect of hydrogen sulfide (H2S) on traumatic brain injury (TBI) induced memory deficits in rats. Brain Res Bull 2017; 134:177-182. [DOI: 10.1016/j.brainresbull.2017.07.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 11/19/2022]
|
21
|
Cartagena CM, Mountney A, Hwang H, Swiercz A, Rammelkamp Z, Boutte AM, Shear DA, Tortella FC, Schmid KE. Subacute Changes in Cleavage Processing of Amyloid Precursor Protein and Tau following Penetrating Traumatic Brain Injury. PLoS One 2016; 11:e0158576. [PMID: 27428544 PMCID: PMC4948774 DOI: 10.1371/journal.pone.0158576] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 06/19/2016] [Indexed: 01/22/2023] Open
Abstract
Traumatic brain injury (TBI) is an established risk factor for the development of Alzheimer's disease (AD). Here the effects of severe penetrating TBI on APP and tau cleavage processing were investigated in a rodent model of penetrating ballistic-like brain injury (PBBI). PBBI was induced by stereotactically inserting a perforated steel probe through the right frontal cortex of the anesthetized rat and rapidly inflating/deflating the probe's elastic tubing into an elliptical shaped balloon to 10% of total rat brain volume causing temporary cavitation injury. Separate animals underwent probe injury (PrI) alone without balloon inflation. Shams underwent craniectomy. Brain tissue was collected acutely (4h, 24h, 3d) and subacutely (7d) post-injury and analyzed by immunoblot for full length APP (APP-FL) and APP beta c-terminal fragments (βCTFs), full length tau (tau-FL) and tau truncation fragments and at 7d for cytotoxic Beta amyloid (Aβ) peptides Aβ40 and Aβ42 analysis. APP-FL was significantly decreased at 3d and 7d following PBBI whereas APP βCTFs were significantly elevated by 4h post-injury and remained elevated through 7d post-injury. Effects on βCTFs were mirrored with PrI, albeit to a lesser extent. Aβ40 and Aβ42 were significantly elevated at 7d following PBBI and PrI. Tau-FL decreased substantially 3d and 7d post-PBBI and PrI. Importantly, a 22 kDa tau fragment (tau22), similar to that found in AD, was significantly elevated by 4h and remained elevated through 7d post-injury. Thus both APP and tau cleavage was dramatically altered in the acute and subacute periods post-injury. As cleavage of these proteins has also been implicated in AD, TBI pathology shown here may set the stage for the later development of AD or other tauopathies.
Collapse
Affiliation(s)
- Casandra M. Cartagena
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Andrea Mountney
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Hye Hwang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Adam Swiercz
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Zoe Rammelkamp
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Angela M. Boutte
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Deborah A. Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Frank C. Tortella
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Kara E. Schmid
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| |
Collapse
|
22
|
Li G, Kim C, Kim J, Yoon H, Zhou H, Kim J. Common Pesticide, Dichlorodiphenyltrichloroethane (DDT), Increases Amyloid-β Levels by Impairing the Function of ABCA1 and IDE: Implication for Alzheimer's Disease. J Alzheimers Dis 2016; 46:109-22. [PMID: 25720399 DOI: 10.3233/jad-150024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
While early-onset familial Alzheimer's disease (AD) is caused by a genetic mutation, the vast majority of late-onset AD is likely caused by the combination of genetic and environmental factors. Unlike genetic studies, potential environmental factors affecting AD pathogenesis have not yet been thoroughly investigated. Among environmental factors, pesticides seem to be one of critical environmental contributors to late-onset AD. Recent studies reported that the serum and brains of AD patients have dramatically higher levels of a metabolite of dichlorodiphenyltrichloroethane (DDT). While these epidemiological studies provided initial clues to the environmental risks potentially contributing to disease pathogenesis, a functional approach is required to determine whether they actually have a causal role in disease development. In our study, we addressed this critical knowledge gap by investigating possible mechanisms by which DDT affects amyloid-β (Aβ) levels. We treated H4-AβPPswe or H4 cells with DDT to analyze its effect on Aβ metabolism using Aβ production, clearance, and degradation assays. We found that DDT significantly increased the levels of amyloid-β protein precursor (AβPP) and β-site AβPP-cleaving enzyme1 (BACE1), affecting Aβ synthesis pathway in H4-AβPPswe cells. Additionally, DDT impaired the clearance and extracellular degradation of Aβ peptides. Most importantly, we identified for the first time that ATP-binding cassette transporter A1 (ABCA1) and insulin-degrading enzyme (IDE) are the downstream target genes adversely affected by DDT. Our findings provide insight into the molecular mechanisms by which DDT exposure may increase the risk of AD, and it further supports that ABCA1 and IDE may be potential therapeutic targets.
Collapse
Affiliation(s)
- Gongbo Li
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China.,Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Chaeyoung Kim
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Jaekwang Kim
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Hyejin Yoon
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, USA.,Neurobiology of Disease Graduate Program, Mayo Graduate School, Jacksonville, FL, USA
| | - Huadong Zhou
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jungsu Kim
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, USA.,Neurobiology of Disease Graduate Program, Mayo Graduate School, Jacksonville, FL, USA
| |
Collapse
|
23
|
Yu S, Li S, Henke A, Muse ED, Cheng B, Welzel G, Chatterjee AK, Wang D, Roland J, Glass CK, Tremblay M. Dissociated sterol-based liver X receptor agonists as therapeutics for chronic inflammatory diseases. FASEB J 2016; 30:2570-9. [PMID: 27025962 DOI: 10.1096/fj.201600244r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/21/2016] [Indexed: 12/26/2022]
Abstract
Liver X receptor (LXR), a nuclear hormone receptor, is an essential regulator of immune responses. Activation of LXR-mediated transcription by synthetic agonists, such as T0901317 and GW3965, attenuates progression of inflammatory disease in animal models. However, the adverse effects of these conventional LXR agonists in elevating liver lipids have impeded exploitation of this intriguing mechanism for chronic therapy. Here, we explore the ability of a series of sterol-based LXR agonists to alleviate inflammatory conditions in mice without hepatotoxicity. We show that oral treatment with sterol-based LXR agonists in mice significantly reduces dextran sulfate sodium colitis-induced body weight loss, which is accompanied by reduced expression of inflammatory markers in the large intestine. The anti-inflammatory property of these agonists is recapitulated in vitro in mouse lamina propria mononuclear cells, human colonic epithelial cells, and human peripheral blood mononuclear cells. In addition, treatment with LXR agonists dramatically suppresses inflammatory cytokine expression in a model of traumatic brain injury. Importantly, in both disease models, the sterol-based agonists do not affect the liver, and the conventional agonist T0901317 results in significant liver lipid accumulation and injury. Overall, these results provide evidence for the development of sterol-based LXR agonists as novel therapeutics for chronic inflammatory diseases.-Yu, S., Li, S., Henke, A., Muse, E. D., Cheng, B., Welzel, G., Chatterjee, A. K., Wang, D., Roland, J., Glass, C. K., Tremblay, M. Dissociated sterol-based liver X receptor agonists as therapeutics for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Shan Yu
- California Institute for Biomedical Research, La Jolla, California, USA
| | - Sijia Li
- California Institute for Biomedical Research, La Jolla, California, USA
| | - Adam Henke
- California Institute for Biomedical Research, La Jolla, California, USA
| | - Evan D Muse
- Department of Cellular and Molecular Medicine and Scripps Translational Science Institute, Scripps Health, La Jolla, California, USA and
| | - Bo Cheng
- California Institute for Biomedical Research, La Jolla, California, USA
| | - Gustav Welzel
- California Institute for Biomedical Research, La Jolla, California, USA
| | | | - Danling Wang
- California Institute for Biomedical Research, La Jolla, California, USA
| | - Jason Roland
- California Institute for Biomedical Research, La Jolla, California, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine and Department of Medicine, University of California, San Diego, La Jolla, California, USA; and
| | - Matthew Tremblay
- California Institute for Biomedical Research, La Jolla, California, USA;
| |
Collapse
|
24
|
Dong X, Liu T, Xu S, Zhu L, Zhang P, Cheng A, Qian Q. The relevance of ABCA1 R219K polymorphisms and serum ABCA1 protein concentration to Parkinson’s disease pathogenesis and classification: a case–control study. Genes Genomics 2016. [DOI: 10.1007/s13258-015-0354-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
25
|
Bird SM, Sohrabi HR, Sutton TA, Weinborn M, Rainey-Smith SR, Brown B, Patterson L, Taddei K, Gupta V, Carruthers M, Lenzo N, Knuckey N, Bucks RS, Verdile G, Martins RN. Cerebral amyloid-β accumulation and deposition following traumatic brain injury--A narrative review and meta-analysis of animal studies. Neurosci Biobehav Rev 2016; 64:215-28. [PMID: 26899257 DOI: 10.1016/j.neubiorev.2016.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 01/15/2016] [Indexed: 10/22/2022]
Abstract
Traumatic brain injury (TBI) increases the risk of neurodegenerative disorders many years post-injury. However, molecular mechanisms underlying the relationship between TBI and neurodegenerative diseases, such as Alzheimer's disease (AD), remain to be elucidated. Nevertheless, previous studies have demonstrated a link between TBI and increased amyloid-β (Aβ), a protein involved in AD pathogenesis. Here, we review animal studies that measured Aβ levels following TBI. In addition, from a pool of initially identified 1209 published papers, we examined data from 19 eligible animal model studies using a meta-analytic approach. We found an acute increase in cerebral Aβ levels ranging from 24h to one month following TBI (overall log OR=2.97 ± 0.40, p<0.001). These findings may contribute to further understanding the relationship between TBI and future dementia risk. The methodological inconsistencies of the studies discussed in this review suggest the need for improved and more standardised data collection and study design, in order to properly elucidate the role of TBI in the expression and accumulation of Aβ.
Collapse
Affiliation(s)
- Sabine M Bird
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Hamid R Sohrabi
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Thomas A Sutton
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Michael Weinborn
- Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia; School of Psychology, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Belinda Brown
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Leigh Patterson
- Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Veer Gupta
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Malcolm Carruthers
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Centre for Men's Health, 96 Harley Street, London, W1G 7HY, United Kingdom
| | - Nat Lenzo
- Oceanic Medical Imaging, Hollywood Medical Centre, 85 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Neville Knuckey
- Centre for Neuromuscular and Neurological Disorders (CNND), University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Romola S Bucks
- School of Psychology, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Giuseppe Verdile
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; School of Biomedical Sciences, CHIRI Biosciences, Curtin University, Kent Street, Bentley, 6102 WA, Australia
| | - Ralph N Martins
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia.
| |
Collapse
|
26
|
Hong S, Washington PM, Kim A, Yang CP, Yu TS, Kernie SG. Apolipoprotein E Regulates Injury-Induced Activation of Hippocampal Neural Stem and Progenitor Cells. J Neurotrauma 2015; 33:362-74. [PMID: 25905575 DOI: 10.1089/neu.2014.3860] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Partial recovery from even severe traumatic brain injury (TBI) is ubiquitous and occurs largely through unknown mechanisms. Recent evidence suggests that hippocampal neural stem/progenitor cell (NSPC) activation and subsequent neurogenesis are responsible for at least some aspects of spontaneous recovery following TBI. Apolipoprotein E (ApoE) regulates postnatal neurogenesis in the hippocampus and is therefore a putative mediator of injury-induced neurogenesis. Further, ApoE isoforms in humans are associated with different cognitive outcomes following TBI. To investigate the role of ApoE in injury-induced neurogenesis, we exposed wild-type, ApoE-deficient, and human ApoE isoform-specific (ApoE3 and ApoE4) transgenic mice crossed with nestin-green fluorescent protein (GFP) reporter mice to controlled cortical impact (CCI) and assessed progenitor activation at 2 d post-injury using unbiased stereology. GFP+ progenitor cells were increased by approximately 120% in the ipsilateral hippocampus in injured wild-type mice, compared with sham mice (p<0.01). Co-localization of GFP+ cells with bromodeoxyrudine (BrdU) to label dividing cells indicated increased proliferation of progenitors in the injured hippocampus (p<0.001). This proliferative injury response was absent in ApoE-deficient mice, as no increase in GFP+ cells was observed in the injured hippocampus, compared with sham mice, despite an overall increase in proliferation indicated by increased BrdU+ cells (86%; p<0.05). CCI-induced proliferation of GFP+ cells in both ApoE3 and ApoE4 mice but the overall response was attenuated in ApoE4 mice due to fewer GFP+ cells at baseline. We demonstrate that ApoE is required for injury-induced proliferation of NSPCs after experimental TBI, and that this response is influenced by human APOE genotype.
Collapse
Affiliation(s)
- Sue Hong
- 1 Departments of Pediatrics and Pathology and Cell Biology, Columbia University College of Physicians and Surgeons , New York, New York
| | - Patricia M Washington
- 1 Departments of Pediatrics and Pathology and Cell Biology, Columbia University College of Physicians and Surgeons , New York, New York
| | - Ahleum Kim
- 1 Departments of Pediatrics and Pathology and Cell Biology, Columbia University College of Physicians and Surgeons , New York, New York
| | - Cui-Ping Yang
- 2 Key Laboratory of Animal Models and Human Disease Mechanisms , Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Tzong-Shiue Yu
- 1 Departments of Pediatrics and Pathology and Cell Biology, Columbia University College of Physicians and Surgeons , New York, New York
| | - Steven G Kernie
- 1 Departments of Pediatrics and Pathology and Cell Biology, Columbia University College of Physicians and Surgeons , New York, New York
| |
Collapse
|
27
|
Ellagic acid prevents cognitive and hippocampal long-term potentiation deficits and brain inflammation in rat with traumatic brain injury. Life Sci 2015; 124:120-7. [PMID: 25637685 DOI: 10.1016/j.lfs.2015.01.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/26/2014] [Accepted: 01/17/2015] [Indexed: 11/24/2022]
Abstract
AIMS Traumatic brain injury (TBI) remains one of the main clinical problems globally and is a common cause of death among youth. Cognitive defects such as thinking, memory and behavior or mental health disorders are considered as the most frequent effects of severe and moderate TBI. It has been reported that ellagic acid (EA), a natural polyphenol, exhibits protective effects against oxidative damage. This study was performed to examine the EA preventive effects on cognitive impairments, long-term potentiation (LTP) deficits in hippocampus and brain inflammation induced by diffuse TBI in rat. MAIN METHODS Subchronic oral administration of 100 mg/kg EA, 7 consecutive days before induction of trauma (once daily) was used to elucidate the EA effects on passive avoidance memory and hippocampal LTP following TBI. To illustrate the possible mechanisms related to the preventive effects of EA on brain function following TBI, brain content of IL-1β, IL-6 and blood-brain barrier (BBB) permeability were determined. KEY FINDINGS EA pretreatment significantly (P<0.001) prevented TBI-induced memory and hippocampal LTP impairments in rat. Furthermore TBI induced elevation in brain content of IL-1β, IL-6 and BBB permeability were decreased significantly (P<0.001) due to EA pre-treatment. SIGNIFICANCE Our findings suggest that EA can prevent cognitive and LTP deficits and also prevent brain inflammation following TBI.
Collapse
|
28
|
Elder GA, Stone JR, Ahlers ST. Effects of low-level blast exposure on the nervous system: is there really a controversy? Front Neurol 2014; 5:269. [PMID: 25566175 PMCID: PMC4271615 DOI: 10.3389/fneur.2014.00269] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 11/29/2014] [Indexed: 12/20/2022] Open
Abstract
High-pressure blast waves can cause extensive CNS injury in human beings. However, in combat settings, such as Iraq and Afghanistan, lower level exposures associated with mild traumatic brain injury (mTBI) or subclinical exposure have been much more common. Yet controversy exists concerning what traits can be attributed to low-level blast, in large part due to the difficulty of distinguishing blast-related mTBI from post-traumatic stress disorder (PTSD). We describe how TBI is defined in human beings and the problems posed in using current definitions to recognize blast-related mTBI. We next consider the problem of applying definitions of human mTBI to animal models, in particular that TBI severity in human beings is defined in relation to alteration of consciousness at the time of injury, which typically cannot be assessed in animals. However, based on outcome assessments, a condition of "low-level" blast exposure can be defined in animals that likely approximates human mTBI or subclinical exposure. We review blast injury modeling in animals noting that inconsistencies in experimental approach have contributed to uncertainty over the effects of low-level blast. Yet, animal studies show that low-level blast pressure waves are transmitted to the brain. In brain, low-level blast exposures cause behavioral, biochemical, pathological, and physiological effects on the nervous system including the induction of PTSD-related behavioral traits in the absence of a psychological stressor. We review the relationship of blast exposure to chronic neurodegenerative diseases noting the paradoxical lowering of Abeta by blast, which along with other observations suggest that blast-related TBI is pathophysiologically distinct from non-blast TBI. Human neuroimaging studies show that blast-related mTBI is associated with a variety of chronic effects that are unlikely to be explained by co-morbid PTSD. We conclude that abundant evidence supports low-level blast as having long-term effects on the nervous system.
Collapse
Affiliation(s)
- Gregory A. Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James R. Stone
- Department of Radiology, University of Virginia, Charlottesville, VA, USA
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, USA
| |
Collapse
|
29
|
Jullienne A, Roberts JM, Pop V, Paul Murphy M, Head E, Bix GJ, Badaut J. Juvenile traumatic brain injury induces long-term perivascular matrix changes alongside amyloid-beta accumulation. J Cereb Blood Flow Metab 2014; 34:1637-45. [PMID: 25052558 PMCID: PMC4269722 DOI: 10.1038/jcbfm.2014.124] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/28/2014] [Accepted: 06/05/2014] [Indexed: 02/08/2023]
Abstract
In our juvenile traumatic brain injury (jTBI) model, emergence of cognitive dysfunctions was observed up to 6 months after trauma. Here we hypothesize that early brain injury induces changes in the neurovascular unit (NVU) that would be associated with amyloid-beta (Aβ) accumulation. We investigated NVU changes for up to 6 months in a rat jTBI model, with a focus on the efflux protein P-glycoprotein (P-gp) and on the basement membrane proteins perlecan and fibronectin, all known to be involved in Aβ clearance. Rodent-Aβ staining is present and increased after jTBI around cerebral blood microvessels, and the diameter of those is decreased by 25% and 34% at 2 and 6 months, respectively, without significant angiogenesis. P-glycoprotein staining in endothelium is decreased by 22% and parallels an increase of perlecan and fibronectin staining around cerebral blood vessels. Altogether, these results strongly suggest that the emergence of long-term behavioral dysfunctions observed in rodent jTBI may be related to endothelial remodeling at the blood-brain barrier alongside vascular dysfunction and altered Aβ trafficking. This study shows that it is important to consider jTBI as a vascular disorder with long-term consequences on cognitive functions.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, Loma Linda University, Loma Linda, California, USA
| | - Jill M Roberts
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Viorela Pop
- Department of Pediatrics, Loma Linda University, Loma Linda, California, USA
| | - M Paul Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Gregory J Bix
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Jérôme Badaut
- 1] Department of Pediatrics, Loma Linda University, Loma Linda, California, USA [2] Department of Physiology, Loma Linda University, Loma Linda, California, USA [3] CNRS UMR 5287, Bordeaux University, Bordeaux, France
| |
Collapse
|
30
|
Gandy S, Ikonomovic MD, Mitsis E, Elder G, Ahlers ST, Barth J, Stone JR, DeKosky ST. Chronic traumatic encephalopathy: clinical-biomarker correlations and current concepts in pathogenesis. Mol Neurodegener 2014; 9:37. [PMID: 25231386 PMCID: PMC4249716 DOI: 10.1186/1750-1326-9-37] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 05/13/2014] [Indexed: 12/14/2022] Open
Abstract
Background Chronic traumatic encephalopathy (CTE) is a recently revived term used to describe a neurodegenerative process that occurs as a long term complication of repetitive mild traumatic brain injury (TBI). Corsellis provided one of the classic descriptions of CTE in boxers under the name “dementia pugilistica” (DP). Much recent attention has been drawn to the apparent association of CTE with contact sports (football, soccer, hockey) and with frequent battlefield exposure to blast waves generated by improvised explosive devices (IEDs). Recently, a promising serum biomarker has been identified by measurement of serum levels of the neuronal microtubule associated protein tau. New positron emission tomography (PET) ligands (e.g., [18 F] T807) that identify brain tauopathy have been successfully deployed for the in vitro and in vivo detection of presumptive tauopathy in the brains of subjects with clinically probable CTE. Methods Major academic and lay publications on DP/CTE were reviewed beginning with the 1928 paper describing the initial use of the term CTE by Martland. Results The major current concepts in the neurological, psychiatric, neuropsychological, neuroimaging, and body fluid biomarker science of DP/CTE have been summarized. Newer achievements, such as serum tau and [18 F] T807 tauopathy imaging, are also introduced and their significance has been explained. Conclusion Recent advances in the science of DP/CTE hold promise for elucidating a long sought accurate determination of the true prevalence of CTE. This information holds potentially important public health implications for estimating the risk of contact sports in inflicting permanent and/or progressive brain damage on children, adolescents, and adults.
Collapse
Affiliation(s)
- Sam Gandy
- Departments of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY 10029, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Leeds PR, Yu F, Wang Z, Chiu CT, Zhang Y, Leng Y, Linares GR, Chuang DM. A new avenue for lithium: intervention in traumatic brain injury. ACS Chem Neurosci 2014; 5:422-33. [PMID: 24697257 DOI: 10.1021/cn500040g] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of disability and death from trauma to central nervous system (CNS) tissues. For patients who survive the initial injury, TBI can lead to neurodegeneration as well as cognitive and motor deficits, and is even a risk factor for the future development of neurodegenerative disorders such as Alzheimer's disease. Preclinical studies of multiple neuropathological and neurodegenerative disorders have shown that lithium, which is primarily used to treat bipolar disorder, has considerable neuroprotective effects. Indeed, emerging evidence now suggests that lithium can also mitigate neurological deficits incurred from TBI. Lithium exerts neuroprotective effects and stimulates neurogenesis via multiple signaling pathways; it inhibits glycogen synthase kinase-3 (GSK-3), upregulates neurotrophins and growth factors (e.g., brain-derived neurotrophic factor (BDNF)), modulates inflammatory molecules, upregulates neuroprotective factors (e.g., B-cell lymphoma-2 (Bcl-2), heat shock protein 70 (HSP-70)), and concomitantly downregulates pro-apoptotic factors. In various experimental TBI paradigms, lithium has been shown to reduce neuronal death, microglial activation, cyclooxygenase-2 induction, amyloid-β (Aβ), and hyperphosphorylated tau levels, to preserve blood-brain barrier integrity, to mitigate neurological deficits and psychiatric disturbance, and to improve learning and memory outcome. Given that lithium exerts multiple therapeutic effects across an array of CNS disorders, including promising results in preclinical models of TBI, additional clinical research is clearly warranted to determine its therapeutic attributes for combating TBI. Here, we review lithium's exciting potential in ameliorating physiological as well as cognitive deficits induced by TBI.
Collapse
Affiliation(s)
- Peter R. Leeds
- Molecular
Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, Maryland 20892-1363, United States
| | - Fengshan Yu
- Molecular
Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, Maryland 20892-1363, United States
| | - Zhifei Wang
- Molecular
Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, Maryland 20892-1363, United States
| | - Chi-Tso Chiu
- Molecular
Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, Maryland 20892-1363, United States
| | | | - Yan Leng
- Molecular
Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, Maryland 20892-1363, United States
| | - Gabriel R. Linares
- Molecular
Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, Maryland 20892-1363, United States
| | - De-Maw Chuang
- Molecular
Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, MSC 1363, Bethesda, Maryland 20892-1363, United States
| |
Collapse
|
32
|
Jasmin SB, Pearson V, Lalonde D, Domenger D, Théroux L, Poirier J. Differential regulation of ABCA1 and ABCG1 gene expressions in the remodeling mouse hippocampus after entorhinal cortex lesion and liver-X receptor agonist treatment. Brain Res 2014; 1562:39-51. [PMID: 24661912 DOI: 10.1016/j.brainres.2014.03.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/14/2014] [Accepted: 03/15/2014] [Indexed: 12/30/2022]
Abstract
Entorhinal cortex lesioning (ECL) causes an extensive deafferentation of the hippocampus that is classically followed by a compensatory reinnervation, where apolipoprotein E, the main extracellular lipid-carrier in the CNS, has been shown to play a crucial role by shuttling cholesterol to reconstructing neurons terminals. Hence, we investigated whether the ATP-binding cassette (ABC) transporters -A1 and -G1, known to regulate cellular cholesterol efflux and lipidation of the apolipoprotein E-containing lipoprotein complex are actively involved in this context of brain׳s plastic response to neurodegeneration and deafferentation. We assessed ABCA1 and ABCG1 mRNA and protein levels throughout the degenerative phase and the reinnervation process and evaluated the associated cholinergic sprouting following ECL in the adult mouse brain. We subsequently tested the effect of the pharmacological activation of the nuclear receptor LXR, prior to versus after ECL, on hippocampal ABCA1 and G1 expression and on reinnervation. ECL induced a time-dependent up-regulation of ABCA1, but not G1, that coincided with a significant increase in acetylcholine esterase (AChE) activity in the ipsilateral hippocampus. Pre-ECL, but not post-ECL i.p. treatment with the LXR agonist TO901317 also led to a significant increase solely in hippocampal ABCA1 expression, paralleled by increases in both AchE and synaptophysin protein levels in the deafferented hippocampus. Thus, ABCA1 and -G1 are differentially regulated in the lesioned brain and upon treatment with an LXR agonist. Further, TO901317-induced up-regulation of ABCA1 appears to be more beneficial in a prevention (pre-lesion) than rescue (post-lesion) treatment; both findings support a central role for ABC transporters in brain plasticity.
Collapse
Affiliation(s)
- Stéphanie Bélanger Jasmin
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H4A 2B4; Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec, Canada H4H 1R3
| | - Vanessa Pearson
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H4A 2B4; Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec, Canada H4H 1R3
| | - Daphnée Lalonde
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H4A 2B4; Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec, Canada H4H 1R3
| | - Dorothée Domenger
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H4A 2B4; Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec, Canada H4H 1R3
| | - Louise Théroux
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec, Canada H4H 1R3
| | - Judes Poirier
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H4A 2B4; Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec, Canada H4H 1R3.
| |
Collapse
|
33
|
Chodobski A, Zink BJ, Szmydynger-Chodobska J. Blood-brain barrier pathophysiology in traumatic brain injury. Transl Stroke Res 2013; 2:492-516. [PMID: 22299022 DOI: 10.1007/s12975-011-0125-x] [Citation(s) in RCA: 439] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The blood-brain barrier (BBB) is formed by tightly connected cerebrovascular endothelial cells, but its normal function also depends on paracrine interactions between the brain endothelium and closely located glia. There is a growing consensus that brain injury, whether it is ischemic, hemorrhagic, or traumatic, leads to dysfunction of the BBB. Changes in BBB function observed after injury are thought to contribute to the loss of neural tissue and to affect the response to neuroprotective drugs. New discoveries suggest that considering the entire gliovascular unit, rather than the BBB alone, will expand our understanding of the cellular and molecular responses to traumatic brain injury (TBI). This review will address the BBB breakdown in TBI, the role of blood-borne factors in affecting the function of the gliovascular unit, changes in BBB permeability and post-traumatic edema formation, and the major pathophysiological factors associated with TBI that may contribute to post-traumatic dysfunction of the BBB. The key role of neuroinflammation and the possible effect of injury on transport mechanisms at the BBB will also be described. Finally, the potential role of the BBB as a target for therapeutic intervention through restoration of normal BBB function after injury and/or by harnessing the cerebrovascular endothelium to produce neurotrophic growth factors will be discussed.
Collapse
Affiliation(s)
- Adam Chodobski
- Neurotrauma and Brain Barriers Research Laboratory, Department of Emergency Medicine, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | | | | |
Collapse
|
34
|
Abstract
Traumatic brain injury (TBI) affects all age groups in a population and is an injury generating scientific interest not only as an acute event, but also as a complex brain disease with several underlying neurobehavioral and neuropathological characteristics. We review early and long-term alterations after juvenile and adult TBI with a focus on changes in the neurovascular unit (NVU), including neuronal interactions with glia and blood vessels at the blood-brain barrier (BBB). Post-traumatic changes in cerebral blood-flow, BBB structures and function, as well as mechanistic pathways associated with brain aging and neurodegeneration are presented from clinical and experimental reports. Based on the literature, increased attention on BBB changes should be integrated in studies characterizing TBI outcome and may provide a meaningful therapeutic target to resolve detrimental post-traumatic dysfunction.
Collapse
Affiliation(s)
- V Pop
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354 USA
| | | |
Collapse
|
35
|
Washington PM, Morffy N, Parsadanian M, Zapple DN, Burns MP. Experimental traumatic brain injury induces rapid aggregation and oligomerization of amyloid-beta in an Alzheimer's disease mouse model. J Neurotrauma 2013; 31:125-34. [PMID: 24050316 DOI: 10.1089/neu.2013.3017] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Soluble amyloid-beta (Aβ) oligomers are hypothesized to be the pathogenic species in Alzheimer's disease (AD), and increased levels of oligomers in the brain subsequent to traumatic brain injury (TBI) may exacerbate secondary injury pathways and underlie increased risk of developing AD in later life. To determine whether TBI causes Aβ aggregation and oligomerization in the brain, we exposed triple transgenic AD model mice to controlled cortical impact injury and measured levels of soluble, insoluble, and oligomeric Aβ by enzyme-linked immunosorbent assay (ELISA) at 1, 3, and 7 days postinjury. TBI rapidly increased levels of both soluble and insoluble Aβ40 and Aβ42 in the injured cortex at 1 day postinjury. We confirmed previous findings that identified damaged axons as a major site of Aβ accumulation using both immunohistochemistry and biochemistry. We also report that soluble Aβ oligomers were significantly increased in the injured cortex, as demonstrated by both ELISA and Western blot. Interestingly, the mouse brain is able to rapidly clear trauma-induced Aβ, with both soluble and insoluble Aβ species returning to sham levels by 7 days postinjury. In conclusion, we demonstrate that TBI causes acute accumulation and aggregation of Aβ in the brain, including the formation of low- and high-molecular-weight Aβ oligomers. The formation and aggregation of Aβ into toxic species acutely after injury may play a role in secondary injury cascades after trauma and, chronically, may contribute to increased risk of developing AD in later life.
Collapse
Affiliation(s)
- Patricia M Washington
- 1 Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center , Washington, DC
| | | | | | | | | |
Collapse
|
36
|
Winston CN, Chellappa D, Wilkins T, Barton DJ, Washington PM, Loane DJ, Zapple DN, Burns MP. Controlled cortical impact results in an extensive loss of dendritic spines that is not mediated by injury-induced amyloid-beta accumulation. J Neurotrauma 2013; 30:1966-72. [PMID: 23879560 DOI: 10.1089/neu.2013.2960] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The clinical manifestations that occur after traumatic brain injury (TBI) include a wide range of cognitive, emotional, and behavioral deficits. The loss of excitatory synapses could potentially explain why such diverse symptoms occur after TBI, and a recent preclinical study has demonstrated a loss of dendritic spines, the postsynaptic site of the excitatory synapse, after fluid percussion injury. The objective of this study was to determine if controlled cortical impact (CCI) also resulted in dendritic spine retraction and to probe the underlying mechanisms of this spine loss. We used a unilateral CCI and visualized neurons and dendtritic spines at 24 h post-injury using Golgi stain. We found that TBI caused a 32% reduction of dendritic spines in layer II/III of the ipsilateral cortex and a 20% reduction in the dendritic spines of the ipsilateral dentate gyrus. Spine loss was not restricted to the ipsilateral hemisphere, however, with similar reductions in spine numbers recorded in the contralateral cortex (25% reduction) and hippocampus (23% reduction). Amyloid-β (Aβ), a neurotoxic peptide commonly associated with Alzheimer disease, accumulates rapidly after TBI and is also known to cause synaptic loss. To determine if Aβ contributes to spine loss after brain injury, we administered a γ-secretase inhibitor LY450139 after TBI. We found that while LY450139 administration could attenuate the TBI-induced increase in Aβ, it had no effect on dendritic spine loss after TBI. We conclude that the acute, global loss of dendritic spines after TBI is independent of γ-secretase activity or TBI-induced Aβ accumulation.
Collapse
Affiliation(s)
- Charisse N Winston
- 1 Department of Neuroscience, Laboratory for Brain Injury and Dementia, Georgetown University Medical Center , Washington, DC
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Walker KR, Tesco G. Molecular mechanisms of cognitive dysfunction following traumatic brain injury. Front Aging Neurosci 2013; 5:29. [PMID: 23847533 PMCID: PMC3705200 DOI: 10.3389/fnagi.2013.00029] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/18/2013] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) results in significant disability due to cognitive deficits particularly in attention, learning and memory, and higher-order executive functions. The role of TBI in chronic neurodegeneration and the development of neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic Lateral Sclerosis (ALS) and most recently chronic traumatic encephalopathy (CTE) is of particular importance. However, despite significant effort very few therapeutic options exist to prevent or reverse cognitive impairment following TBI. In this review, we present experimental evidence of the known secondary injury mechanisms which contribute to neuronal cell loss, axonal injury, and synaptic dysfunction and hence cognitive impairment both acutely and chronically following TBI. In particular we focus on the mechanisms linking TBI to the development of two forms of dementia: AD and CTE. We provide evidence of potential molecular mechanisms involved in modulating Aβ and Tau following TBI and provide evidence of the role of these mechanisms in AD pathology. Additionally we propose a mechanism by which Aβ generated as a direct result of TBI is capable of exacerbating secondary injury mechanisms thereby establishing a neurotoxic cascade that leads to chronic neurodegeneration.
Collapse
Affiliation(s)
- Kendall R Walker
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine Boston, MA, USA
| | | |
Collapse
|
38
|
Loren J, Huang Z, Laffitte BA, Molteni V. Liver X receptor modulators: a review of recently patented compounds (2009 - 2012). Expert Opin Ther Pat 2013; 23:1317-35. [PMID: 23826715 DOI: 10.1517/13543776.2013.814640] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION The development of small molecule agonists of the liver X receptors (LXRs) has been an area of interest for over a decade, given the critical role of those receptors in cholesterol metabolism, glucose homeostasis, inflammation, innate immunity and lipogenesis. Many potential indications have been characterized over time including atherosclerosis, diabetes, inflammation, Alzheimer's disease and cancer. However, concerns about the lipogenic effects of full LXRα/β agonists have required extensive efforts aimed at identifying LXRβ agonist with limited activity on the LXRα receptor to increase the safety margins. AREAS COVERED This review includes a summary of the LXR agonists that have reached the clinic and summarizes the patent applications for LXR modulators from September 2009 to December 2012 with emphasis on chemical matters, biological data associated with selected analogs and therapeutic indications. EXPERT OPINION As LXR agonists have the potential to be useful for many indications, the scientific community, despite setbacks due to on-target side effects, has maintained interest and devised strategies to overcome safety hurdles. While a clinical proof of concept still remains elusive, the recent advancement of compounds into the clinic highlights that acceptable safety margins in preclinical species have been achieved.
Collapse
Affiliation(s)
- Jon Loren
- Genomics Institute of the Novartis Research Foundation , 10675 John Jay Hopkins Drive, San Diego, CA 92121 , USA +001 858 332 4736 ;
| | | | | | | |
Collapse
|
39
|
Shackleford G, Makoukji J, Grenier J, Liere P, Meffre D, Massaad C. Differential regulation of Wnt/beta-catenin signaling by Liver X Receptors in Schwann cells and oligodendrocytes. Biochem Pharmacol 2013; 86:106-14. [DOI: 10.1016/j.bcp.2013.02.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 02/27/2013] [Accepted: 02/27/2013] [Indexed: 11/16/2022]
|
40
|
Abrahamson EE, Foley LM, Dekosky ST, Hitchens TK, Ho C, Kochanek PM, Ikonomovic MD. Cerebral blood flow changes after brain injury in human amyloid-beta knock-in mice. J Cereb Blood Flow Metab 2013; 33:826-33. [PMID: 23443172 PMCID: PMC3677107 DOI: 10.1038/jcbfm.2013.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Traumatic brain injury (TBI) is an environmental risk factor for Alzheimer's disease (AD). Increased brain concentrations of amyloid-β (Aβ) peptides and impaired cerebral blood flow (CBF) are shared pathologic features of TBI and AD and promising therapeutic targets. We used arterial spin-labeling magnetic resonance imaging to examine if CBF changes after TBI are influenced by human Aβ and amenable to simvastatin therapy. CBF was measured 3 days and 3 weeks after controlled cortical impact (CCI) injury in transgenic human Aβ-expressing APP(NLh/NLh) mice compared to murine Aβ-expressing C57Bl/6J wild types. Compared to uninjured littermates, CBF was reduced in the cortex of the injured hemisphere in both Aβ transgenics and wild types; deficits were more pronounced in the transgenic group, which exhibited injury-induced increased concentrations of human Aβ. In the hemisphere contralateral to CCI, CBF levels were stable in Aβ transgenic mice but increased in wild-type mice, both relative to uninjured littermates. Post-injury treatment of Aβ transgenic mice with simvastatin lowered brain Aβ concentrations, attenuated deficits in CBF ipsilateral to injury, restored hyperemia contralateral to injury, and reduced brain tissue loss. Future studies examining long-term effects of simvastatin therapy on CBF and chronic neurodegenerative changes after TBI are warranted.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Álvarez XA, Figueroa J, Muresanu D. Peptidergic drugs for the treatment of traumatic brain injury. FUTURE NEUROLOGY 2013. [DOI: 10.2217/fnl.12.95] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Traumatic brain injury (TBI) is a devastating medical condition that has an enormous socioeconomic impact because it affects more than 10 million people annually worldwide and is associated with high rates of hospitalization, mortality and disability. Although TBI survival has improved continuously for decades, particularly in developing countries, implementation of an effective drug therapy for TBI represents an unmet clinical need. All confirmatory trials conducted to date with drugs targeting a single TBI pathological pathway failed to show clinical efficacy, probably because TBI pathophysiology involves multiple cellular and molecular mechanisms of secondary brain damage. According to current scientific evidence of the participation of peptide-mediated mechanisms in the processes of brain injury and repair after TBI, peptidergic drugs represent a multimodal therapy alternative to improve acute outcome and long-term recovery in TBI patients. Preliminary randomized-controlled clinical trials and open-label studies conducted to date with the peptidergic drug Cerebrolysin® (Ever Neuro Pharma GmbH, Unterach, Austria) and with the endogenous neuropeptides progesterone and erythropoietin, showed positive clinical results. Cerebrolysin-treated patients had a faster clinical recovery, a shorter hospitalization time and a better long-term outcome. Treatment with progesterone showed advantages over placebo regarding TBI mortality and clinical outcome, whereas erythropoietin only reduced mortality. Further validation of these promising findings in confirmatory randomized-controlled clinical trials is warranted. This article reviews the scientific basis and clinical evidence on the development of multimodal peptidergic drugs as a therapeutic option for the effective treatment of TBI patients.
Collapse
Affiliation(s)
| | - Jesús Figueroa
- Rehabilitation Department, Santiago University Hospital, Santiago de Compostela, Spain
| | - Dafin Muresanu
- Department of Neurology, University of Medicine & Pharmacy ‘Iuliu Hatieganu’, Cluj-Napoca, Romania
| |
Collapse
|
42
|
Namjoshi DR, Martin G, Donkin J, Wilkinson A, Stukas S, Fan J, Carr M, Tabarestani S, Wuerth K, Hancock REW, Wellington CL. The liver X receptor agonist GW3965 improves recovery from mild repetitive traumatic brain injury in mice partly through apolipoprotein E. PLoS One 2013; 8:e53529. [PMID: 23349715 PMCID: PMC3547922 DOI: 10.1371/journal.pone.0053529] [Citation(s) in RCA: 41] [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: 09/15/2012] [Accepted: 11/29/2012] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) increases Alzheimer’s disease (AD) risk and leads to the deposition of neurofibrillary tangles and amyloid deposits similar to those found in AD. Agonists of Liver X receptors (LXRs), which regulate the expression of many genes involved in lipid homeostasis and inflammation, improve cognition and reduce neuropathology in AD mice. One pathway by which LXR agonists exert their beneficial effects is through ATP-binding cassette transporter A1 (ABCA1)-mediated lipid transport onto apolipoprotein E (apoE). To test the therapeutic utility of this pathway for TBI, we subjected male wild-type (WT) and apoE−/− mice to mild repetitive traumatic brain injury (mrTBI) followed by treatment with vehicle or the LXR agonist GW3965 at 15 mg/kg/day. GW3965 treatment restored impaired novel object recognition memory in WT but not apoE−/− mice. GW3965 did not significantly enhance the spontaneous recovery of motor deficits observed in all groups. Total soluble Aβ40 and Aβ42 levels were significantly elevated in WT and apoE−/− mice after injury, a response that was suppressed by GW3965 in both genotypes. WT mice showed mild but significant axonal damage at 2 d post-mrTBI, which was suppressed by GW3965. In contrast, apoE−/− mice showed severe axonal damage from 2 to 14 d after mrTBI that was unresponsive to GW3965. Because our mrTBI model does not produce significant inflammation, the beneficial effects of GW3965 we observed are unlikely to be related to reduced inflammation. Rather, our results suggest that both apoE-dependent and apoE-independent pathways contribute to the ability of GW3965 to promote recovery from mrTBI.
Collapse
Affiliation(s)
- Dhananjay R. Namjoshi
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Georgina Martin
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - James Donkin
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna Wilkinson
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Sophie Stukas
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jianjia Fan
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Carr
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Sepideh Tabarestani
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kelli Wuerth
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert E. W. Hancock
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail: *
| |
Collapse
|
43
|
De Gasperi R, Gama Sosa MA, Kim SH, Steele JW, Shaughness MC, Maudlin-Jeronimo E, Hall AA, Dekosky ST, McCarron RM, Nambiar MP, Gandy S, Ahlers ST, Elder GA. Acute blast injury reduces brain abeta in two rodent species. Front Neurol 2012; 3:177. [PMID: 23267342 PMCID: PMC3527696 DOI: 10.3389/fneur.2012.00177] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 12/02/2012] [Indexed: 12/02/2022] Open
Abstract
Blast-induced traumatic brain injury (TBI) has been a major cause of morbidity and mortality in the conflicts in Iraq and Afghanistan. How the primary blast wave affects the brain is not well understood. In particular, it is unclear whether blast injures the brain through mechanisms similar to those found in non-blast closed impact injuries (nbTBI). The β-amyloid (Aβ) peptide associated with the development of Alzheimer’s disease is elevated acutely following TBI in humans as well as in experimental animal models of nbTBI. We examined levels of brain Aβ following experimental blast injury using enzyme-linked immunosorbent assays for Aβ 40 and 42. In both rat and mouse models of blast injury, rather than being increased, endogenous rodent brain Aβ levels were decreased acutely following injury. Levels of the amyloid precursor protein (APP) were increased following blast exposure although there was no evidence of axonal pathology based on APP immunohistochemical staining. Unlike the findings in nbTBI animal models, levels of the β-secretase, β-site APP cleaving enzyme 1, and the γ-secretase component presenilin-1 were unchanged following blast exposure. These studies have implications for understanding the nature of blast injury to the brain. They also suggest that strategies aimed at lowering Aβ production may not be effective for treating acute blast injury to the brain.
Collapse
Affiliation(s)
- Rita De Gasperi
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center Bronx, NY, USA ; Department of Psychiatry, Mount Sinai School of Medicine New York, NY, USA ; Friedman Brain Institute, Mount Sinai School of Medicine New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Reiss AB, Voloshyna I. Regulation of cerebral cholesterol metabolism in Alzheimer disease. J Investig Med 2012; 60:576-82. [PMID: 22367100 DOI: 10.2310/jim.0b013e318246d973] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alzheimer disease (AD) is an age-related neurodegenerative disorder that manifests as a progressive loss of memory and deterioration of higher cognitive functions. Alzheimer disease is characterized by accumulation in the brain of the β-amyloid peptide generated by β- and γ-secretase processing of amyloid precursor protein. Epidemiological studies have linked elevated plasma cholesterol and lipoprotein levels in midlife with AD development. Cholesterol-fed animal models exhibit neuropathologic features of AD including accumulation of β-amyloid peptide. Specific isoforms of the cholesterol transporter apolipoprotein E are associated with susceptibility to AD. Although multiple lines of evidence indicate a role for cholesterol in AD, the exact impact and mechanisms involved remain largely unknown. This review summarizes the current state of our knowledge of the influence of cholesterol and lipid pathways in AD pathogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Allison B Reiss
- Winthrop Research Institute, Department of Medicine, Winthrop University Hospital, Mineola, NY 11501, USA.
| | | |
Collapse
|
45
|
Maulik M, Westaway D, Jhamandas JH, Kar S. Role of cholesterol in APP metabolism and its significance in Alzheimer's disease pathogenesis. Mol Neurobiol 2012; 47:37-63. [PMID: 22983915 DOI: 10.1007/s12035-012-8337-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/19/2012] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disorder believed to be initiated by accumulation of amyloid β (Aβ)-related peptides derived from proteolytic processing of amyloid precursor protein (APP). Research over the past two decades provided a mechanistic link between cholesterol and AD pathogenesis. Genetic polymorphisms in genes regulating the pivotal points in cholesterol metabolism have been suggested to enhance the risk of developing AD. Altered neuronal membrane cholesterol level and/or subcellular distribution have been implicated in aberrant formation, aggregation, toxicity, and degradation of Aβ-related peptides. However, the results are somewhat contradictory and we still do not have a complete understanding on how cholesterol can influence AD pathogenesis. In this review, we summarize our current understanding on the role of cholesterol in regulating the production/function of Aβ-related peptides and also examine the therapeutic potential of regulating cholesterol homeostasis in the treatment of AD pathology.
Collapse
Affiliation(s)
- M Maulik
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | | | | | | |
Collapse
|
46
|
Yu F, Zhang Y, Chuang DM. Lithium reduces BACE1 overexpression, β amyloid accumulation, and spatial learning deficits in mice with traumatic brain injury. J Neurotrauma 2012; 29:2342-51. [PMID: 22583494 PMCID: PMC3430485 DOI: 10.1089/neu.2012.2449] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Traumatic brain injury (TBI) leads to both acute injury and long-term neurodegeneration, and is a major risk factor for developing Alzheimer's disease (AD). Beta amyloid (Aβ) peptide deposits in the brain are one of the pathological hallmarks of AD. Aβ levels increase after TBI in animal models and in patients with head trauma, and reducing Aβ levels after TBI has beneficial effects. Lithium is known to be neuroprotective in various models of neurodegenerative disease, and can reduce Aβ generation by modulating glycogen synthase kinase-3 (GSK-3) activity. In this study we explored whether lithium would reduce Aβ load after TBI, and improve learning and memory in a mouse TBI model. Lithium chloride (1.5 mEq/kg, IP) was administered 15 min after TBI, and once daily thereafter for up to 3 weeks. At 3 days after injury, lithium attenuated TBI-induced Aβ load increases, amyloid precursor protein (APP) accumulation, and β-APP-cleaving enzyme-1 (BACE1) overexpression in the corpus callosum and hippocampus. Increased Tau protein phosphorylation in the thalamus was also attenuated after lithium treatment following TBI at the same time point. Notably, lithium treatment significantly improved spatial learning and memory in the Y-maze test conducted 10 days after TBI, and in the Morris water maze test performed 17-20 days post-TBI, in association with increased hippocampal preservation. Thus post-insult treatment with lithium appears to alleviate the TBI-induced Aβ load and consequently improves spatial memory. Our findings suggest that lithium is a potentially useful agent for managing memory impairments after TBI or other head trauma.
Collapse
Affiliation(s)
- Fengshan Yu
- Section on Molecular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Yumin Zhang
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - De-Maw Chuang
- Section on Molecular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
47
|
Abstract
Although the precise cause of Alzheimer's disease is not known, the β-amyloid peptide chains of 40-42 amino acids are suspected to contribute to the disease. The β-amyloid precursor protein is found on many types of cell membranes, and the action of secretases (β and γ) on this precursor protein normally releases the β-amyloids at a high rate into the plasma and the cerebrospinal fluid. However, the concentrations of the β-amyloids in the plasma and the spinal fluid vary considerably between laboratories. The β-amyloids adsorb in the nanomolar concentration range to receptors on neuronal and glial cells. The β-amyloids are internalized, become folded in the β-folded or β-pleated shape, and then stack on each other to form long fibrils and aggregates known as plaques. The β-amyloids likely act as monomers, dimers, or multimers on cell membranes to interfere with neurotransmission and memory before the plaques build up. Treatment strategies include inhibitors of β- and γ-secretase, as well as drugs and physiological compounds to prevent aggregation of the amyloids. Several immune approaches and a cholesterol-lowering strategy are also being tested to remove the β-amyloids.
Collapse
Affiliation(s)
- Philip Seeman
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
48
|
Traumatic brain injury, microglia, and Beta amyloid. Int J Alzheimers Dis 2012; 2012:608732. [PMID: 22666622 PMCID: PMC3359797 DOI: 10.1155/2012/608732] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 03/02/2012] [Indexed: 01/30/2023] Open
Abstract
Recently, there has been growing interest in the association between traumatic brain injury (TBI) and Alzheimer's Disease (AD). TBI and AD share many pathologic features including chronic inflammation and the accumulation of beta amyloid (Aβ). Data from both AD and TBI studies suggest that microglia play a central role in Aβ accumulation after TBI. This paper focuses on the current research on the role of microglia response to Aβ after TBI.
Collapse
|
49
|
Abstract
Alzheimer disease (AD) is an age-related neurodegenerative disorder that manifests as a progressive loss of memory and deterioration of higher cognitive functions. Alzheimer disease is characterized by accumulation in the brain of the β-amyloid peptide generated by β- and γ-secretase processing of amyloid precursor protein. Epidemiological studies have linked elevated plasma cholesterol and lipoprotein levels in midlife with AD development. Cholesterol-fed animal models exhibit neuropathologic features of AD including accumulation of β-amyloid peptide. Specific isoforms of the cholesterol transporter apolipoprotein E are associated with susceptibility to AD. Although multiple lines of evidence indicate a role for cholesterol in AD, the exact impact and mechanisms involved remain largely unknown. This review summarizes the current state of our knowledge of the influence of cholesterol and lipid pathways in AD pathogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Allison B Reiss
- Winthrop Research Institute, Department of Medicine, Winthrop University Hospital, Mineola, NY 11501, USA.
| | | |
Collapse
|
50
|
Corrigan F, Vink R, Blumbergs PC, Masters CL, Cappai R, van den Heuvel C. Characterisation of the effect of knockout of the amyloid precursor protein on outcome following mild traumatic brain injury. Brain Res 2012; 1451:87-99. [PMID: 22424792 DOI: 10.1016/j.brainres.2012.02.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 01/17/2012] [Accepted: 02/19/2012] [Indexed: 01/01/2023]
Abstract
The amyloid precursor protein (APP) increases following traumatic brain injury (TBI), although the functional significance of this remains unclear largely because the functions of the subsequent APP metabolites are so different: Aβ is neurotoxic whilst sAPPα is neuroprotective. To investigate this further, APP wildtype and knockout mice were subjected to mild diffuse TBI and their outcomes compared. APP knockout mice displayed significantly worse cognitive and motor deficits, as demonstrated by the Barnes Maze and rotarod respectively, than APP wildtype mice. This was associated with a significant increase in hippocampal and cortical cell loss, as well as axonal injury, in APP knockout mice and an impaired neuroreparative response as indicated by diminished GAP-43 immunoreactivity when compared to APP wildtype mice. This study is the first to demonstrate that endogenous APP is beneficial following mild TBI, suggesting that the upregulation of APP observed following injury is an acute protective response.
Collapse
Affiliation(s)
- Frances Corrigan
- Discipline of Anatomy and Pathology, School of Medical Sciences, University of Adelaide, Adelaide SA, Australia
| | | | | | | | | | | |
Collapse
|